<<

Supplementary material Vasc Neurol

Supplemental Material Liu Page 1

Supplemental Material

Chinese Stroke Association Guidelines for Clinical Management of

Cerebrovascular Disorders

Executive Summary and 2019 Update of Clinical Management of Ischemic

Cerebrovascular Diseases

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 2

Section 1 Definitions associated with ischemic cerebrovascular diseases.

The definitions of ischemic cerebrovascular disease are shown in Table 1.

Section 2 Emergency assessment and diagnosis of ischemic stroke patients

Please refer to Figure 1 for details of the management process for ischemic stroke

patients in the acute phase.

I. First assessment of emergency

(I) Medical history collection

Acute ischemic stroke (AIS) patients suffer from acute onset. It is very important to

inquire about the time of occurrence of symptoms, onset characteristics and progress,

including symptom persistence, fluctuations and relief. If the disease starts onset

during sleep, the time when the patient's last normal performance should be recorded.

These patients may also get sick shortly before waking up, and the onset time is

uncertain, but clinically the final normal time should still be calculated as the last

normal time [1-2]. Using MRI to screen wake-up stroke patients who are not suitable

for thrombectomy, intravenous helps patients get good prognosis [3].

Before the occurrence of current symptoms, some patients will have similar TIA

symptoms that can be relieved automatically. The time window for thrombolytic

therapy is calculated based on the occurrence time of current symptoms. The inquiry

about whether there are causes related to haemorrhagic stroke before the occurrence

of symptoms, such as emotional excitement, intense exercise, sudden change of body

position, excessive blood pressure reduction, as well as whether there is a history of

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 3

stroke or similar stroke-like diseases (such as epilepsy and hypoglycaemia attacks),

has certain significance for diagnosis. Other medical history should include the

presence or absence of atherosclerosis risk factors (such as hypertension, diabetes,

and hyperlipidaemia) and heart disease risk factors (such as atrial fibrillation), drug

abuse, migraine, infection, trauma, surgery and pregnancy history, and attention

should be paid to the patient's recent , especially the presence or absence of

drugs. For patients with consciousness disorder, doctors can ask their

family members or witnesses about the disease.

(II) Physical examination

After emergency doctors and stroke team doctors evaluate the airway and respiratory

and circulatory functions individually or jointly, they will immediately carry out

general physical examination, including auscultation of large cervical vessels and

heart.

(III) Auxiliary examination (such as blood glucose, electrocardiogram, troponin,

chest radiography)

Timely imaging examination is very important for quick assessment and diagnosis of

patients with possible ischemic stroke. All patients should first select urgent cranial

CT examination to exclude cerebral haemorrhage, and MRI may be chosen if

conditions permit, but should not delay thrombolysis therapy. For patients suspected

of macrovascular occlusion, MRA or CTA should be performed as soon as possible (at

the same time of thrombolysis) to determine whether intravascular treatment

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 4

conditions are available. For the process of skull imagological examination for

patients suspected of ischemic stroke after entering the emergency department, see

Supplemental Table 1.

For all suspected stroke patients, routine laboratory examination should be carried out

immediately upon arrival at the emergency department, including blood glucose, renal

function, electrolyte, blood routine including platelet count, coagulation function

including international standardized ratio (INR), myocardial markers, and

bedside electrocardiogram examination should be performed. Because time is of the

utmost importance, thrombolytic therapy should not be delayed due to waiting for the

above laboratory results, unless the patient has oral or obvious history

of coagulation. Retrospective analysis of patients receiving intravenous thrombolysis

has found that the probability of unexpected coagulation disorders and

thrombocytopenia is very low [4-5].For the vast majority of patients, blood glucose test

results must be acquired before thrombolytic therapy [6].

Some laboratory tests can be selected for specific patients, toxicological screening can

be performed for suspected drug abuse, chest radiography can be performed for

suspected aortic dissection, electroencephalogram can be performed for suspected

epilepsy, lumbar puncture can be performed for suspected intracranial infection or

subarachnoid haemorrhage, blood gas examination can be performed for hypoxia, and

pregnancy tests can be performed for women of childbearing age.

For intravenous thrombolysis or mechanical thrombectomy, the earlier treatment is

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 5

started, the greater the benefit. Many studies abroad suggest that the average or

median door-to-imaging time under different hospital configurations is less than

20min [7-10].However, a large-scale registration study in China in 2011 showed that the

average door-to-needle time (DNT) in China was 116min, and the average imaging-

to-needle time (INT) was 90min. Therefore, based on China's national conditions and

actual clinical practice, skull imagological examination should be completed within

30min for patients suspected of ischemic stroke [11].

Recommendations

[1] It is recommended that patients suspected of having an ischemic stroke

should complete finish brain imaging within 30 minutes of arrival at the

emergency department (Class I, Level of Evidence B).

[2] Emergency assessment of blood glucose, renal function, electrolytes, complete

blood count (including platelet count), blood coagulation (including INR),

cardiac injury markers, and a bedside 12-lead ECG is recommended, but should

not delay the initiation of IV rt-PA. For most patients, only the assessment of

blood glucose must precede the initiation of IV rt-PA (Class I, Level of Evidence

B).

(IV) Definitions of penumbra

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 6

The hypoperfusion brain tissue is divided into three parts according to function and

prognosis. The centre is the irreversible infarction core, and the outermost periphery is

the hypoperfusion area with intact function. The area between them is the ischemic

penumbra. The brain tissue in the penumbra is at risk of irreversible ischemic damage,

but this part of brain tissue can be saved if blood perfusion is restored in time.

Therefore, ischemic penumbra is currently an important therapeutic target for AIS [12].

The early studies mainly explored the best time window to save the penumbra [13-14],

but the general and narrow time window will cause many potentially beneficial

patients to have poor prognosis due to failure to receive . With the

development of multi-mode imaging, the penumbra can be accurately identified by

judging whether the infarct core matches its peripheral hypoperfusion areas, which

prolongs the treatment time window and has important clinical guiding value.

Researchers further set a target mismatch (TMM) by comparing the relationship

between different mismatch degrees and prognosis, which means that more ischemic

penumbra can be saved under a specific mismatch threshold and the prognosis is

better after reperfusion therapy [15]. Common imaging strategies include diffusion-

perfusion imaging mismatch, CTP, clinical imaging mismatch and diffusion-flair

mismatch. Other technologies such as DWI-SWI mismatch, ASL-PWI mismatch,

APTW and MR OMI have been reported, but have not been applied to large-scale

clinical trials [16-19].

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 7

In many large-scale clinical studies, for intravenous thrombolysis or intravascular

therapy, such as DEFUSE, DEFUSE2, EPITHET, DEDAS, DAWN, and DEFUSE3,

the most widely used and mature imaging strategy is diffusion-perfusion imaging

mismatch (DWI-PWI mismatch) [15, 20-22]. It uses the mismatch between DWI

infarction core area and PWI hypoperfusion area to identify ischemic penumbra. At

present, the generally accepted set threshold is: the area with Tmax > 6s on PWI is set

as the hypoperfusion area, the area with ADC < 600×10-6mm2/s on DWI is set as the

infarction core area, and the target mismatch is defined as the area with infarction

core area < 70ml, ischemic penumbra > 15ml, and the area with the ratio of total

hypoperfusion area to ischemic core area being > 1.8 [15].At present, required data can

be directly obtained by using software such as Rapid. Although the DWI-PWI

mismatch imaging strategy can accurately identify ischemic penumbra and provide

important information for clinical reperfusion therapy, it is difficult to promote it in

most clinical centres due to the time-consuming and labour-intensive defects of

emergency MRI.

Another commonly-used method is CTP. It has been widely used in multiple large-

scale clinical trials such as ESCAPE, CRISP, SWIFT PRIME, EXTEND-IA and

DEFUSE 3 [15, 23-25]. At present, the generally accepted set threshold is: the area with

cerebral blood flow (CBF) of the affected side lower than 30% of normal tissue is set

as the infarction core lesion, the area with Tmax > 6s is set as the hypoperfusion area,

and the definition of target mismatch is the same as before. However, setting CBF <

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 8

30% as the infarction core in this imaging strategy is not as accurate as DWI, and

more research is needed to evaluate it. Considering that emergency CT is currently

most widely used and the examination time is short, CTP may have better clinical

operability.

The 2018 DAWN study effectively extended the time window for intravascular

treatment to 24h based on DWI/CTP-clinical mismatch. The researchers divided the

patients into three groups according to the size, age and NIHSS score of the imaging

lesion: Group A was over 80 years old, NIHSS ≥ 10 and infarction core < 21ml. Group

B was under 80 years old, NIHSS ≥ 10 and infarction core < 31ml. Group C was under

80 years old, NIHSS ≥ 20 and infarction core of 31--51ml. Finally, the clinical trial

was terminated in advance due to the obvious benefits of patients in the intravascular

treatment group. Therefore, the imaging strategy is promising, but it still needs to be

verified by more clinical practice [26].

The DWI-FLAIR mismatch strategy is often applied to patients with wake-up stroke

or unclear onset time, such as PRE-FLAIR, WAKE-UP and other studies. DWI-

FLAIR mismatch is defined as lesions visible on DWI but no high signal in

corresponding parts on FLAIR [27]. Researchers believe that DWI-FLAIR mismatch

may indicate stroke that occurs within 4.5h hours. In the WAKE-UP study, the

intravenous thrombolysis group had better clinical prognosis, but it also increased the

risk of intracranial haemorrhage [28].

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 9

Supplemental Table 2 summarizes the set thresholds of different imaging strategies

for ischemic penumbra. At present, some studies have pointed out that it is not

accurate to determine ischemic penumbra with a specific threshold, and proposed a

multi-parameter analysis model that includes imaging and clinical data, that makes

use of CTP, NIHSS score and age as parameters for algorithm analysis, or selects

DWI and PWI parameters, NIHSS score and age as parameters for analysis [29].A MR

RESCUE study concluded through the algorithm model that patients after intravenous

thrombolysis therapy, whether or not with ischemic penumbra, fail to benefit from

intravascular treatment [30].

Recommendations

If feasible, patients with AIS within 6 to 24 hours of last known normal who have

LVO in the anterior circulation, obtaining CTP or DWI with MRI perfusion is

recommended to aid in patient selection for endovascular therapy. Patient

selected for endovascular therapy should be follow the same eligibility criteria of

the two major RCTs (DAWN and DEFUSE 3) (Class IIa, Level of Evidence B).

1. Stroke CT and MRI PWI

(1) Previous RCT studies, including DIAS, DIAS-II, DIAS-III, DEDAS, DEFUSE,

EPITHET and ITAIS-II study in China, used multi-mode images (CTP, diffusion-

perfusion mismatch, , etc.) to extend the time window to screen patients

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 10

who could receive intravenous thrombolysis, but most of the results were not

beneficial. At present, some clinical trials (such as ECASS-IV) are being carried out

to evaluate the guiding value of multi-mode imaging for the time window of

intravenous thrombolysis [31-39].

(2) Six large RCTs, including MR CLEAN, ESCAPE, EXTEND-IA, SWIFT-PRIME,

REVASCAT and THRACE, proved that patients with large artery occlusion can

benefit from intravascular treatment within a time window of 6 hours [25,40-

44].Although multi-mode images (including CTP or MRI) are used to establish the

inclusion criteria in the four RCTs of REVASCAT, SWIFT, EXTEND-IA and SWIFT-

Prime, only non-enhanced CT is used to screen patients with large artery occlusion in

the THRACE and MR CLEAN studies. Therefore, it is believed that in the 6-hour

time window, the use of multi-mode images to screen patients may exclude some

patients with benefits, and may also delay the timing of treatment. However, multi-

mode imaging can evaluate infarct focus, ischemic penumbra and collateral

circulation, which helps guide further treatment. More high-quality RCTs are needed

to evaluate its advantages and disadvantages in the future.

(3) In 2017, the DAWN study combined clinical the NIHSS score and CTP or DWI to

indicate the infarct volume, and established the clinical-image mismatch criteria. The

patients suspected of anterior circulation artery occlusion were screened in a 6-24

hour time window, and were divided into intravascular treatment group and traditional

treatment control group. The 90d function evaluation of the intravascular treatment

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 11

was superior to that of the control group and had statistical significance (mRS score

0--2 , 49% vs. 13%, adjusted difference 33%, 95% CI: 21--44, posterior probability

advantage > 0.999), so the clinical trial was terminated earlier [26].

In 2018, the DEFUSE 3 study used CTP or DWI-PWI to establish the imaging

standard of infarction-ischemia hypoperfusion mismatch. Patients with suspected

anterior circulation artery occlusion were screened in a 6-16 hour time window and

randomly divided into an intravascular treatment group and a traditional treatment

group, and the trial was terminated in advance because the prognosis of the

intravascular treatment group was significantly better than that of the control group

(mRS score 0-2 , 44.6% vs. 16.7%,RR=2.67, 95% CI: 1.60-4.48, P< 0.0001) [45].

The subgroup analysis using the DAWN inclusion criteria also confirmed the benefits.

The above two studies confirmed that intravascular treatment can produce benefits

beyond the time window of 6 hours. Therefore, patients should be screened strictly

according to their inclusion criteria in clinical practice. In the future, more high-

quality RCT is needed to explore indications for intravascular treatment beyond 6

hours.

(4) The ASPECTS score can be used to identify suspected patients with large artery

stroke, create conditions for intravascular treatment, and can also be used to evaluate

treatment prognosis, collateral circulation, etc. [46-50]. At present, the most commonly

used ASPECTS score is based on CT. ASPECTS < 6 was used as the exclusion

criterion in the ESCAPE and SWIFT-PRIME studies, and ASPECTS < 7 was used as

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 12

the exclusion criterion in the REVASCAT study [41, 42]. However, in the MR CLEAN

study, there was no strict restriction on the ASPECTS score. The results showed that

patients in groups 5--7 and 8--10 had no difference in benefits, and patients with 0--4

had no obvious benefits, but they cannot be used as exclusion criteria [51]. A 2017

retrospective study in China compared the clinical prognosis of patients with

ASPECTS 5 and ASPECTS 6 of anterior circulation artery occlusion after

intravascular treatment, and concluded that intravascular treatment could not achieve

satisfactory efficacy for patients with ASPECTS 5 [52]. Some existing studies in China

use ASPECTS ≥ 6 as the inclusion criteria [53]. In conclusion, ASPECTS ≥ 6 is

currently used as one of the inclusion criteria for intravascular treatment, and more

clinical studies are needed to verify the efficacy and safety of intravascular treatment

in people with ASPECTS < 6. At present, ASPECTS scores based on CTP, CTA,

MRI, etc. have become available one after another, and their accuracy, consistency

and applicability need to be studied through more clinical trials [53-57].

(5) At present, most of the imaging studies on AIS are clinical trials based on

intravenous thrombolysis or intravascular treatment, and there is no study specifically

targeting imaging available. More imaging studies based on acute stroke may be

needed in the future.

Recommendations

[1] It is unclear whether using multimodal imaging criteria to select ischemic

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 13

stroke patients who have unclear time of symptom onset for treatment with IV

rt-PA is beneficial or not, therefore is not recommended outside a clinical trial

(Class III, Level of Evidence B).

[2] If needed, multimodal imaging should be obtained as quickly as possible, to

not delay administration of IV rt-PA (Class IIb, Level of Evidence B).

[3] It is unclear whether using perfusion imaging (CTP or PWI) for selecting

patients for endovascular treatment < 6 hours is beneficial (Class IIb, Level of

Evidence B).

[4] It is recommended for AIS patients meeting the eligibility criteria of the two

major RCTs (DAWN and DEFUSE 3) within 6 to 24 hours of last known normal

who have LVO in the anterior circulation to obtain CTP or DWI with MRI

perfusion with subsequent endovascular therapy (Class IIa, Level of Evidence

B).

[5] It is recommended that the ASCPECTS score based on head CT be

considered when evaluating for endovascular treatment. However, the decision-

making doctor must have received the training in the assessment of NIHSS and

ASPECTS scores and been verified for consistency (Class IIa, Level of Evidence

B).

(V) Stroke severity and score

Neurological examination should be brief and comprehensive, but thrombolytic

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 14

therapy should not be delayed. A stroke scale can be used. The National Institute of

Health Stroke Scale (NIHSS) is currently the most commonly used scale in the world

to evaluate neurological deficits of patients. It is easy and simple to operate, takes a

short time, and its clinical application effect has been confirmed [58-59].

Recommendations

For patients with acute presentation of neurological dysfunction, medical history

taking and physical examination must be performed rapidly. Medical history

includes onset characteristics, predisposing factors, last known normal time, past

medical history and current medication list. Physical examination includes vital

signs and general physical examination. The use of NIHSS as a stroke severity

rating scale is recommended (Class I, Level of Evidence A).

II. Emergency diagnosis and differential diagnosis

(I) Diagnostic criteria of AIS

1. In case of acute onset, the specific time of onset or the last normal time is traceable

(onset during sleep).

2. Focal neurological impairment (weakness or numbness of one side of the face or

limbs, language disorders, visual disorders, etc.), a few of which are full-face

neurological impairment.

3. Imaging shows responsible ischemic lesions, or symptoms/signs last for more than

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 15

24 hours.

4. Non-vascular causes are excluded.

5. Cerebral haemorrhage is excluded by head CT/MRI.

(II) Differential diagnosis of stroke mimics

Some non-ischemic cerebrovascular diseases can also show AIS-like symptoms,

which are commonly mental factors, epilepsy, hypoglycaemia, migraine, hypertensive

encephalopathy, central nervous system infection, brain tumour, multiple sclerosis,

electrolyte disturbance, drug toxicity, etc., and should be further identified based on

medical history, clinical manifestations, laboratory examination and imaging

characteristics.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 16

Section III Reperfusion therapy for acute ischemic stroke

I. Patients within 4.5h hours of onset - intravenous thrombolysis treatment

A number of clinical trials have evaluated the efficacy and safety of rt-PA intravenous

thrombolysis for acute cerebral infarction. The therapeutic time window of the study

includes within 3 hours, 3--4.5 hours and 6 hours after onset. The NINDS trial result

showed that the number of patients with complete or nearly complete neurological

function recovery in 3 months in the rt-PA intravenous thrombolysis group was

significantly higher than that in the placebo group within 3 hours, and the mortality of

the two groups was similar [60]. The incidence of symptomatic intracranial

haemorrhage was higher in the intravenous thrombolysis group than in the placebo

group. The ECASS Ⅲ trial showed that intravenous rt-PA was still effective 3--4.5

hours after onset [61]. The IST-3 trial indicated that the benefit of intravenous

thrombolysis with rt-PA within 6 hours of onset is still unclear[62]. The following

systematic review analyzed 12 rt-PA intravenous thrombolysis trial which included

7,012 patients, suggesting that rt-PA intravenous thrombolysis can increase the good

clinical outcomes of patients within 6 hours of onset, and that rt-PA intravenous

thrombolysis within 3 hours of onset has similar effects for patients aged above and

under 80 years old [63].

For patients with slight or rapid improvement of stroke symptoms, who have received

major surgery in the past 3 months and suffered from myocardial infarction recently,

the risk-benefit ratio of intravenous thrombolysis still needs to be weighed and further

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 17

studied. Rt-PA intravenous or arterial thrombolysis may be unfavorable for patients

taking direct inhibitors or direct factor Xa inhibitors, and therefore is not

recommended, unless sensitive laboratory tests such as APTT, INR, platelet count,

Ecarin Clotting Time (ECT), thrombin time (TT) or direct Xa factor activity test are

normal, or these drugs have not been taken for more than 2 days (assuming normal

renal function).

The use of multi-mode MRI or CT to help choose patients whose onset has exceeded

4.5 hour but still with penumbra and who can receive thrombolysis is still a research

hotspot. In terms of the use of rt-PA, in addition to the risk of haemorrhage, partial

obstruction of respiratory tract due to hematogenous oedema has also been reported.

The flow chart of intravenous thrombolysis management for ischemic stroke patients

within 4.5h hours of onset is shown in Figure 2.

(I) Indications and contraindications for intravenous thrombolysis

(Supplemental Table 3 and 4)

(II) Thrombolytic treatment

Basic vital functions (including body temperature, pulse, respiration, blood pressure

and state of consciousness) should be closely monitored. Emergency, including

intracranial hypertension, severe blood pressure abnormality, blood glucose

abnormality, body temperature abnormality, and epilepsy must be handled promptly.

1. Breathing and oxygen inhalation

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 18

(1) Oxygen should be inhaled when necessary. Blood oxygen saturation should be

maintained above 94%. Patients with severe airway dysfunction should be given

airway support (tracheal intubation or incision) and assisted respiration.

(2) Patients without hypoxemia do not need regular oxygen inhalation.

2. Electrocardiogram monitoring and treatment of cardiac lesions

Electrocardiogram examination should be performed routinely within 24 hours after

cerebral infarction. Depending on illness conditions, continuous ECG monitoring

shall be carried out for 24 hours or more when conditions permit, so as to find

paroxysmal atrial fibrillation or severe arrhythmia and other cardiac diseases early.

Drugs that increase heart burden should be avoided or be used with caution.

3. Body temperature control

(1) For patients with elevated body temperature, the causes of fever should be found

and handled. If there is infection, antibiotic treatment should be given.

(2) Antipyretic measures should be taken for patients with body temperature > 38℃.

4. Blood pressure control related to thrombolytic therapy

A number of clinical randomized trials show that the blood pressure of acute stroke

patients should be controlled at systolic pressure < 180mmHg and diastolic pressure <

100mmHg before receiving intravenous thrombolysis treatment, and the

blood pressure of patients should be guaranteed to be < 180/100mmHg within 24

hours after thrombolytic therapy. For AIS patients considering thrombolytic therapy,

special blood pressure management guidelines have been determined (Supplemental

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 19

Table 5), which include reducing blood pressure to below 185/110mmHg in a relaxing

way so as to be suitable for intravenous rt-PA thrombolytic therapy. Once intravenous

administration is given, blood pressure must be maintained at 180/105mmHg to limit

the risk of cerebral haemorrhage. In the first 24 hours, the higher the blood pressure,

the greater the risk of accompanying cerebral haemorrhage, with a linear relationship

between the two. Some observational studies show that stroke patients with high

blood pressure level [64-70] and high blood pressure variability [71] have higher bleeding

risks after receiving Alteplase thrombolytic therapy. The exact blood pressure value

that may lead to high-risk haemorrhagic transformation events after thromboembolic

events is unclear. In the future RCT on intravenous thrombolysis treatment, the blood

pressure level needs to be taken as the research target of concern.

Of the 6 clinical effects RCT (REVASCAT, SWIFT PRIME, EXTEND-IA, THRACE,

ESCAPE, and MR CLEAN) of intravenous thrombolysis bridging intravascular

mechanical thrombectomy and balloon dilatation within 6 hours after stroke, [40-44]) 5

excluded patients with blood pressure levels greater than 185/110mmHg. In another

study (ESCAPE study) [25], there were no exclusion criteria for blood pressure level.

The DAWN study also took > 180/100mmHg as one of its exclusion criteria

[26].Although these RCTs do not clearly explain the reason for setting this blood

pressure value, it is reasonable to recommend this value as a guideline at present

according to their research results.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 20

Recommendations

[1] Patients with elevated blood pressure and other aspects suitable for

intravenous rt-PA therapy should be cautious in lowering blood pressure before

thrombolysis. The recommended goal systolic blood pressure is <180 mm Hg and

diastolic blood pressure is <105 mm Hg (Class I, Level of Evidence B).

[2] It is reasonable to maintain blood pressure (≤180/100 mm Hg) before intra-

arterial therapy in patients who do not receive IV thrombolysis (Class II, Level

of Evidence B).

[3] Within 24 hours after IV R t-PA thrombolysis, blood pressure should be

<180/105 mm Hg (Class I, Level of Evidence B).

5. Blood glucose control

(1) Hyperglycaemia: About 40% of patients have hyperglycemia after stroke, which is

unfavourable for prognosis. Insulin therapy should be given when blood glucose

exceeds l0 mmol/l. Blood glucose monitoring should be strengthened, and the blood

glucose level can be controlled at 7.7--10 mmol/L.

(2) Hypoglycaemia: When blood glucose is lower than 3.3mmol/L, 10%--20%

glucose can be given orally or intravenously. The goal is to reach normal blood

glucose levels.

(III) rt-PA administration dose and method

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 21

Rt-PA 0.9mg/kg (the maximum dose is 90mg) is injected intravenously, of which 10%

is injected intravenously within the first 1min, and the rest 90% is dissolved in 100ml

of normal saline for continuous intravenous drip of 1 hour. Patients should be closely

monitored during medication and within 24 hours of medication (Supplemental Table

6).

(IV) Other recommendations

Age, baseline NIHSS, vascular recanalization and sICH are the main factors affecting

prognosis. The observational meta-analysis showed that compared with patients aged

< 80 years, AIS patients aged > 80 years have 50% less chance of obtaining a good

prognosis. However, another meta-analysis on six RCTs showed that intravenous rt-

PA thrombolysis was still beneficial compared with no thrombolysis in AIS patients

aged > 80 years with the onset < 3h (OR = 1.4, 95% CI: 1.3--1.6, n=3439).

Advanced age and high stroke severity at baseline are important predictors of poor

prognosis. However, according to IST-3 research and meta-analysis, the benefits of rt-

PA intravenous thrombolysis for severe AIS at baseline have not decreased. In the

analysis after the NINDS study, it was found that intravenous thrombolysis for mild

stroke can still produce benefits when the onset is < 3 h [72-73],and meanwhile, the

risk of cerebral infarction haemorrhage conversion is low.

The EXCHANTED study did not prove that the low dose (0.6mg/kg) rt-PA and

standard dose (0.9mg/kg) are equally effective (OR=1.09, 95% CI: 0.95--1.25), but

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 22

the risk of bleeding is reduced (OR=0.48, 95% CI: 0.27--0.86) [74].

The proportion of thrombocytopenia in the population is about 0.3%. The proportion

of INR abnormality is about 0.4% in patients who have not taken and have

no history of suspected coagulation mechanism abnormalities such as end-stage renal

disease and tumour [75].The sample size of intravenous rt-PA thrombolysis study for

patients with low molecular weight was small, but the results showed that the

sICH risk increases (OR=8.4, 95% CI: 2.2--32.2), death risk increases (OR=5.3, 95%

CI: 1.8--15.5), and the proportion of good prognosis decreases by 68% [76].

The observational study shows that baseline MRI indicates microbleeds and increased

sICH risk after intravenous rt-PA thrombolysis (RR=2.36, 95% CI: 1.21--4.61). When

the number of microbleeds is > 10, the risk of sICH is significantly increased

(RR=7.01, 95% CI: 3.20--15.38). Since the proportion of microbleeds > 10 is less than

1%, it is not recommended to perform conventional MRI before intravenous

thrombolysis to exclude microbleeds [77].

Evidence for the combined use of Ⅱb/Ⅲa receptor antagonists such as and

rt-PA intravenous thrombolysis is insufficient.[78].The retrospective case-control study

shows that early therapy within 24 hours after intravenous

thrombolysis may be safe, and the risk of haemorrhage is not increased.

Recommendations

Recommendation Class of Recommendation

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 23

Level of Evidence

Within 3 hours of onset, rt-PA IV thrombolytic therapy is Class I, Level of Evidence A

recommended for patients aged over 18 and who meet other

criteria

For patients who are suitable for IV thrombolysis within 3 Class I, Level of Evidence A

hours of onset, rt-PA IV thrombolysis is recommended (drug

dose 0.9 mg/kg, maximum dose 90 mg, continuous infusion

within 60 minutes, of which 10% of the first dose is IV infusion

within 1 minute)

For AIS patients with severe symptoms within 3 hours of onset, Class I, Level of Evidence A

rt-PA IV thrombolysis is recommended. Although the risk of

bleeding events increases, it still benefits

For patients with mild symptoms but with disabling stroke Class I, Level of Evidence B

symptoms within 3 hours of onset, rt-PA IV thrombolytic

therapy is recommended. Current studies have shown that rt-

PA IV thrombolytic therapy is beneficial for these patients

rt-PA IV thrombolysis is still recommended for patients Class I, Level of Evidence B

suitable for IV thrombolysis within 3-4.5 hours of onset

The benefit of rt-PA thrombolytic therapy for AIS patients aged Class IIb, Level of Evidence B

over 80 years within 3-4.5 hours after onset is not clear

Within 4.5 hours of AIS onset, low dose IV rt-PA can be given Class IIb, Level of Evidence B

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 24

to patients with potential high risk of haemorrhagic events.

Usage: rt-PA 0.6 mg/kg (maximum dose is 60 mg), of which

15% of the total amount was intravenously injected within the

first 1 minute, and the remaining 85% was intravenously

infused with infusion pump for 1 hour

Considering the low incidence of platelet abnormalities and Class IIa, Level of Evidence B

coagulation dysfunction in the general population, IV

thrombolysis should not be delayed while waiting for the results

of platelet counts when there is no reason to suspect that the

results of the tests are abnormal

The safety and efficacy of intravenous rt-PA therapy in AIS Class III, Level of Evidence C

patients with potential bleeding risk or coagulation disorders

have not been determined the safety and efficacy of IV rt-PA

therapy in AIS patients with potential haemorrhagic risk or

coagulation disorders have not been determined

IV rt-PA is not suitable for patients who have used low Class III, Level of Evidence B

molecular weight heparin within 24 hours, regardless

prophylactic or therapeutic dose

Pre-thrombolytic MRI examination showed that IV Class IIa, Level of Evidence B

thrombolysis was reasonable in patients with a number of (1-

10) cerebral microbleeds

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 25

Pre-thrombolytic MRI examination showed that IV Class IIa, Level of Evidence B

thrombolysis was associated with an increased risk of

symptomatic intracerebral hemorrhage in patients with a

number of (>10) cerebral microbleeds, and the clinical benefit

is not clear. If there may be significant potential benefits, IV

thrombolysis may be reasonable

During IV thrombolysis, physicians should be fully prepared to Class I, Level of Evidence B

respond to emergency adverse reactions, including

hemorrhagic complications and vasogenic edema that may

cause airway obstruction

Abciximab cannot be used concurrently with IV rt-PA Class III, Level of Evidence B

Whether or not the endovascular treatment is bridged, the risk Class IIb, Level of Evidence B

of starting antiplatelet therapy within 24 h after IV

thrombolysis remains unclear.

In some specific cases, there is a lack of high-level evidence-based medical evidence

such as RCT or cohort studies in terms of the benefits or risks of thrombolysis.

However, there is consensus among most experts as follows for reference.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 26

Recommendation

Recommendation Class of Recommendation

Level of Evidence

The time from onset to treatment has a major impact on prognosis, and rt-PA IV thrombolysis cannot be postponed to Class IIa, Level of Evidence C

wait if symptoms are relieved

For patients with mild non-disabling AIS, IV rt-PA therapy may be suitable within 3 hours of onset Class IIa, Level of Evidence C

IV rt-PA may be beneficial for AIS patients who had a history of digestive tract or urinary bleeding Class IIa, Level of Evidence C

AIS IV thrombolysis may be considered within 14 days after surgery, but should weigh the benefit of thrombolysis and Class IIb, Level of Evidence C

the risk of hemorrhage of the surgical site

AIS patients with recent major trauma history (within 14 days, without affecting the head), physician should carefully Class IIb, Level of Evidence C

consider IV rt-PA treatment and should weigh the risk of the wound hemorrhage and the severity of stroke and

subsequent disability

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 27

The safety and efficacy of IV rt-PA therapy in patients with AIS who have a history of vascular perforation within 7 Class IIb, Level of Evidence C

days are not known

AIS patients with lumbar puncture within 7 days, the safety of IV rt-PA therapy is uncertain Class IIb, Level of Evidence C

AIS patients with abnormal baseline glucose [< 50 mg/dl (2.78 mmol/L) or > 400 mg/dl (22.2 mmol/L)], followed by Class IIb, Level of Evidence C

normalized blood glucose, the benefit of IV rt-PA is not determined

AIS patients with convulsions may benefit from IV rt-PA therapy if there is evidence that limb dysfunction is due to Class IIa, Level of Evidence C

stroke rather than paralysis after seizures

AIS patients who have a known or suspected extracranial carotid artery dissection with an onset time <4.5 h, IV rt-PA Class IIa, Level of Evidence C

therapy should be chosen carefully

AIS patients who have a known or suspected intracranial carotid artery dissection, the efficacy and safety of IV rt-PA Class IIb, Level of Evidence C

therapy has not been established

AIS patients with a small or moderate (<10 mm) unruptured intracranial aneurysm, IV rt-PA therapy may be Class IIa, Level of Evidence C

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 28

considered may be considered

AIS patients with large unruptured or unstable intracranial aneurysms, the risk and effectiveness of IV rt-PA Class IIb, Level of Evidence C

thrombolysis is uncertain

AIS patients with unruptured or untreated intracranial vascular malformations, the safety and risk of IV rt-PA therapy Class IIb, Level of Evidence C

is not known

AIS patients with neuroectodermal tumours may benefit from IV rt-PA thrombolysis Class IIa, Level of Evidence C

AIS with acute myocardial infarction may be considered for intravenous thrombolysis according to the appropriate rt- Class IIa, Level of Evidence C

PA dose of AIS, followed by PCI or stent for acute coronary syndrome AIS patients with acute myocardial infarction

may be considered for IV thrombolysis with the appropriate rt-PA dose of AIS, followed by PCI or stent for acute

coronary syndrome

AIS patients with recent myocardial infarction (>3 months), rt-PA thrombolysis may be beneficial if there is also a non- Class IIa, Level of Evidence C

ST elevation myocardial infarction, or ST elevation myocardial infarction involving the right ventricle/inferior wall

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 29

AIS combined with recent myocardial infarction (>3 months), if ST elevated, and involving the left ventricle/anterior Class IIb, Level of Evidence C

wall, the safety and risk of IV rt-PA thrombolysis is uncertain

Severe AIS with acute pericarditis which may lead to severe disability (mRS score 3 to 5 points), the benefit of IV rt-PA Class IIb, Level of Evidence C

thrombolysis is not clear. An urgent cardiologist consultation is required

Mild or moderate AIS with acute pericarditis or left atrial/ventricular thrombus, the risk and benefit of IV rt-PA Class III, Level of Evidence C

thrombolysis is unknown

Severe AIS with left atrial/ventricular thrombus, or atrial myxoma, or papillary fibroids, may have severe disability Class IIb, Level of Evidence C

(mRS score 3 to 5 points). The safety and efficacy of IV rt-PA are unknown

AIS with cardiovascular or cerebrovascular DSA, IV rt-PA thrombolysis may be benefit. Patients should be carefully Class IIa, Level of Evidence A

assessed for indications, contraindications, and relative contraindications

The efficacy and safety of IV rt-PA thrombolysis in AIS patients with malignancy is unknown. If the expected survival Class IIb, Level of Evidence C

period is >6 months, no other contraindications, no coagulation abnormalities or bleeding, careful consideration of IV

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 30

rt-PA thrombolysis is suitable

Pregnant women with moderate or severe stroke may benefit from intravenous rt-PA thrombolysis if the benefits of Class IIa, Level of Evidence C

intravenous thrombolysis outweigh the risk of uterine bleeding.

The evidence of benefit and risk of IV rt-PA thrombolysis for AIS patients within 14 days postpartum is insufficient Class IIb, Level of Evidence C

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 31

(V) Other thrombolytic drugs

In addition to rt-PA, different studies have evaluated the efficacy and safety of

intravenous thrombolysis with (UK), , TNK-tPA and

ultrasound-assisted intravenous thrombolysis respectively [81]. A study shows that

thrombolysis with urokinase is relatively safe and effective in AIS within 6 hours of

onset [32]. In the desmoteplase study (DIAS-3), after the AIS patients with severe

stenosis or macrovascular occlusion 3--9 hours after onset were given desmoteplase

(90 μg/kg) or placebo, there was no statistical difference in efficacy and safety

between them. Desmoteplase did not improve the efficacy or increase the proportion

of bleeding [98]. In the ultrasound-assisted intravenous thrombolysis study (NOR-

SASS), the AIS patients within 4.5h hours of onset were given TNK-tPA or rt-PA

intravenous thrombolysis, and then randomly divided into the treatment group or

placebo group. There was no statistical difference in the efficacy endpoint (24h

NIHSS improvement rate and 90d neurological function outcome) or safety endpoint

(sICH) between the two groups [79].

Recommendations

[1] Urokinase is safe for those who are not suitable for rt-PA treatment within 6

hours of onset. However, the validity needs further confirmation by high quality

large sample size RCT (Class IIb, Level of Evidence B).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 32

[2] It has not been confirmed that IV injection of thrombolysis

(single dose of 0.4 mg/kg) is superior or inferior to rt-PA. However, for patients

with mild neurological dysfunction without occlusion of the intracranial artery,

tenecteplase can be considered instead of rt-PA (Class IIb, Level of Evidence B).

[3] In addition to clinical trials, ultrasound thrombolysis is not recommended as

an adjunct therapy to IV thrombolysis. Desmoplatinolytic thrombolysis is not

recommended under imaging guidance (Class III, Level of Evidence B).

II. Patients within 6 hours of onset-bridging/intravascular treatment

In the PROACT-II study, the patients with middle cerebral artery (M1 or M2)

occlusion within 6 hours of onset were given recombinant prourokinase combined

with heparin arterial thrombolysis (experimental group) or heparin-only arterial

thrombolysis (control group) respectively. The proportion of 3-month good

neurological function prognosis (mRS score 0--2) in the primary endpoint of the

experimental group was higher than that in the control group (40% vs. 25%, P =

0.04). The recanalization rate of middle cerebral artery (MCA) in the experimental

group was higher than that in the control group (66% vs. 18%, P< 0.001). However,

the incidence rate of symptomatic cerebral haemorrhage in the experimental group

was higher than that in the control group (10% vs. 2%, P=0.06), and the mortality rate

of the two groups was similar.

The MELT trial compared the effect of drug therapy (treatment group) and urokinase

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 33

therapy for artery within 6 hours. The incidence rate of good neurological prognosis

(mRS score 0--2) in 3 months in the treatment group was higher than that in the

control group (49.1% vs. 36.8%, P=0.35). The overall treatment effect and the

incidence rate of symptomatic cerebral haemorrhage were consistent with PROACT-

II trial [80,81].

The early exploratory experiment used a small sample study to evaluate the efficacy

of intravenous low-dose rt-PA combined with arterial thrombolysis. According to the

research results of Emergency Management of Stroke (EMS), Interventional

Management Study I (IMS I) and Interventional Management Study II (IMS II), the

neurological function prognosis of the combined treatment group was significantly

better than that of the control group [82-84].The Basilar Artery International

Cooperation Study (BASICS) reviewed and analysed the clinical effects of

antithrombotic therapy, intravenous thrombolytic therapy or arterial thrombolytic

therapy for patients with acute onset of basilar artery occlusion, and did not show

significant differences among various therapeutic schemes [85].

The recent intravascular trial suggested that arterial thrombolysis has limited effect,

but it can also be used as salvage therapy instead of main therapy. Among 141

patients in the THRACE trial intervention group, 15 patients (11%) used alteplase in

artery after mechanical thrombectomy, with an average dosage of 8.8mg. Compared

with mechanical thrombectomy alone, it had no effect on clinical prognosis [44].

Bridging therapy refers to intra-arterial interventional reperfusion therapy based on

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 34

intravenous thrombolysis, which is divided into direct bridging therapy and salvage

bridging therapy. Direct bridging refers to direct thrombectomy without observing and

waiting for thrombolytic effect after intravenous thrombolysis. Salvage bridging

refers to observing changes in neurological function of patients after intravenous

thrombolysis, and further considering thrombectomy after failure of intravenous

thrombolysis.

At present, intravenous thrombolysis is the first choice for patients within the

intravenous thrombolysis time window. In five randomized controlled studies of early

thrombectomy, more than 90% of patients were treated with mechanical

thrombectomy bridging therapy based on intravenous thrombolysis. For the clinical

effects of bridging therapy and direct thrombectomy, randomized controlled trials are

underway. The subgroup analysis of the HERMES study shows that there is no

difference in prognosis between patients treated with bridging therapy (n=1090) and

patients treated with direct thrombectomy (n=188) (P=0.43), but most patients treated

with direct thrombectomy are due to contraindications for intravenous thrombolysis

[86]. SWIFT, STAR and other non-randomized controlled trials also suggest that direct

thrombectomy and bridging therapy have similar results. Intravenous thrombolysis

before mechanical thrombectomy does not improve clinical benefits compared with

mechanical thrombectomy alone [87-89]. However, some studies suggest that bridging

therapy has good prognosis, low mortality rate, high recanalization rate, less times of

thrombectomy, shorter thrombectomy time and no increase in sICH risk [90-93].

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 35

In the recently published EXTEND-IA TNK study, after intravenous thrombolysis

with TNK-tP before bridging therapy, the good reperfusion rate before thrombectomy

can reach 22% for AIS patients with macrovascular occlusion within 4.5h hours of

onset, which is significantly higher than the 10% reperfusion rate of alteplase

(difference 12%, 95% CI: 2--21; incidence ratio: 2.2, 95% CI: 1.1--4.4; Non-

inferiority P = 0.002; optimal efficiency P=0.03). There is no difference in sICH

between the two groups, and the good prognosis of the TNK-tPA group is better after

90 days. The study suggests that the new intravenous thrombolytic drugs can increase

the recanalization ratio of macrovascular occlusion before thrombectomy, thus

achieve early recovery of perfusion and improvement of prognosis [94].

Mechanical thrombectomy device has received extensive attention because of its

many theoretical advantages: rapid recanalization, lower haemorrhage conversion rate

and prolonged stroke intervention time window [95]. The Food and Drug

Administration (FDA) approved MERCI Retrieval™ (2004) and Penumbra Stroke

Systems™ (2008) as the first generation of mechanical thrombectomy devices. In

2013, three trials were conducted to evaluate the treatment of AIS with intravascular

mechanical thrombectomy- Interventional Management Study III (IMS III),

Mechanical Retrieval and Recanalization of Stroke Clots Using Embolectomy (MR

RESCUE) and A Randomized Controlled Trial on Intra-arterial and Intravenous

Thrombolysis in Acute Ischemic Stroke (SYNTHESIS EXPANSION), all of which

reported negative results [96-98], and failed to manifest the advantages of intravascular

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 36

treatment. This may be due to the following reasons: there was a long delay between

the occurrence of symptoms and treatment, the adopted imaging method failed to

screen out the potential benefited population, the recanalization rate was lower than

expected, and the older generation of thrombectomy equipment was applied.

The US FDA approved the Solitaire™ and Trevo™ stent thrombectomy devices in

2012. After confirming the efficacy and safety of the new-generation stent

thrombectomy device, a number of randomized controlled trials with the new-

generation stent thrombectomy device as the main equipment have confirmed the

advantages of thrombectomy therapy compared with intravenous thrombolysis or

drug therapy alone in patients with macrovascular occlusion. Since the end of 2014, a

series of related studies have successively released relatively consistent research

results: among the screened patients with anterior circulation macrovascular AIS,

intravascular treatment centering on mechanical thrombectomy can bring definite

benefits.

According to the results of 6 randomized controlled trials (MR CLEAN, SWIFT

PRIME, EXTEND-IA, ESCAPE, REVASCAT, THRACE) of mechanical

thrombectomy centring on recyclable stents, the 2015 guidelines give the highest-

level recommendations for thrombectomy to specific populations [44].

Recommendations

[1] Mechanical thrombectomy is strongly recommended for patients within 6h

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 37

after AIS if they meet all the following criteria: (1) pre-stroke mRS score of 0 to

1; (2) causative occlusion of the internal carotid artery or MCA segment 1 (M1);

(3) age ≥18 years; (4) NIHSS score of ≥6; and (5) ASPECTS of ≥6. (Class I, Level

of Evidence A)

[2] It is reasonable to initial treatment with intra-arterial thrombolysis within 6h

after AIS caused by occlusions of the MCA for carefully selected patients who

have contraindications or no clinical response to the use of IV rt-PA and couldn’t

perform mechanical thrombectomy (Class IIa, Level of Evidence B).

[3] Endovascular treatment should be performed as soon as possible after its

indication. Patients eligible for IV rt-PA should receive IV rt-PA and direct

perform bridging treatment for mechanical thrombectomy (Class I, Level of

Evidence A).

[4] Mechanical thrombectomy should performed as the first line treatment for

patients who have contraindications to the use of IV rt-PA (Class IIa, Level of

Evidence A).

[5] Stent retrievers is indicated for mechanical thrombectomy as first choice

(Class I, Level A). Other thrombectomy or aspiration devices approved by local

health authorities may be used at the operators’ discretion (Class IIa, Level of

Evidence B).

[6] Mechanical thrombectomy with stent retrievers may be reasonable for

carefully selected patients with AIS in whom treatment can be initiated (groin

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 38

puncture) within 6 hours of symptom onset and who have causative occlusion of

the MCA segment 2 (M2) or MCA segment 3 (M3) portion of the MCAs (Class

IIb, Level of Evidence B).

[7] Mechanical thrombectomy with stent retrievers may be reasonable for

carefully selected patients with AIS in whom treatment can be initiated (groin

puncture) within 6 hours of symptom onset and who have causative occlusion of

the anterior cerebral arteries, vertebral arteries, basilar artery, or posterior

cerebral arteries (Class IIb, Level of Evidence C).

[8] Mechanical thrombectomy with stent retrievers may be reasonable for

patients with AIS in whom treatment can be initiated (groin puncture) within 6

hours of symptom onset and who have pre-stroke mRS score >1, ASPECTS <6,

or NIHSS score <6, and causative occlusion of the internal carotid artery (ICA)

or proximal MCA (M1). Additional randomized trial data are needed (Class IIb,

Level of Evidence B).

III. Patients within 6--24 hours of onset-intravascular treatment

Progress has been made in the research on mechanical thrombectomy since 2015. The

publication of DWI or CTP Assessment with Clinical Mismatch in the Triage of

Wake-Up and Late Presenting Undergoing Neurointervention with Trevo

(DAWN) and DEFUSE 3 (Endovascular Therapy Following Imaging Evaluation for

Ischemic Stroke 3) has extended the time window for mechanical thrombectomy from

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 39

6 hours to 24 hours. The results of these two randomized controlled trials suggest that

intravascular treatment with stent thrombectomy is still very effective for patients

with acute stroke over 6 hours of onset after screening suitable patients through

preoperative evaluation. To sum up, there is clear evidence to support the use of AIS

intravascular therapy. The success rate and recanalization rate of different mechanical

thrombectomy devices are high in randomized controlled trials using a new

generation of thrombectomy devices. Accurate patient selection schemes and efficient

reperfusion therapy technology are the keys for patients with acute macrovascular

occlusion to benefit from intravascular therapy.

Recommendations

[1] In selected patients with AIS within 6 to 16 hours of last known normal who

have LVO in the anterior circulation and meet other DAWN or DEFUSE 3

eligibility criteria, mechanical thrombectomy is recommended (Class I, Level of

Evidence A).

[2] In selected patients with AIS within 16 to 24 hours of last known normal who

have LVO in the anterior circulation and meet other DAWN eligibility criteria,

mechanical thrombectomy is reasonable (Class IIa, Level of Evidence B).

[3] Patients with acute basilar artery occlusion within 6 to 24 hours should be

evaluated in centers with multimodal imaging and treated with mechanical

thrombectomy or they may be treated within a randomized controlled trial for

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 40

thrombectomy approved by the local ethical committee (Class IIb, Level of

Evidence B).

[4] The benefits of mechanical thrombectomy are uncertain for patients with

acute large vascular occlusion for more than 24 hours (Class IIb, Level of

Evidence C)

See Supplemental Table 7 for a summary of imaging screening criteria beyond time

windows in different studies.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 41

Section 4 Anti-platelet aggregation therapy for acute ischemic cerebrovascular

disease

I. Monotherapy against platelet aggregation

(I)

Two large-scale clinical trials have confirmed that aspirin can significantly reduce the

mortality or disability rate at the end of follow-up, reduce recurrence, and only

slightly increase the risk of symptomatic intracranial haemorrhage [99, 100]. A meta-

analysis of 41283 ischemic stroke patients from 8 clinical studies shows that 160-300

mg aspirin within 48 hours of stroke onset can significantly reduce the risk of early

stroke recurrence and achieve better long-term prognosis, and is not the main risk

factor for early haemorrhage complications [101]. Another meta-analysis comparing

aspirin with placebo shows that aspirin for long-term secondary prevention and

treatment can significantly reduce the risk of stroke (haemorrhagic or ischemic stroke)

by 15% [102].

There is still controversy about the ideal therapeutic dose of aspirin. A study from

Canada believes that the dose of aspirin should be 130 mg/d [103]. In the UK-TIA trial

in the UK, 300mg/d aspirin is as effective as higher doses [104]. In a low-dose aspirin

study in Sweden, 75mg/d aspirin can reduce the incidence of stroke and mortality by

18%, with statistical significance [105]. At present, most neurologists recommend a

dose of 50-325 mg/d.

The subgroup analysis of the NINDS study shows that thrombolysis effect is better

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 42

and incidence of adverse prognosis is lower for patients taking aspirin in the early

phase, but the incidence of sICH is not statistically significant compared with that of

patients not taking aspirin in the early phase [106]. Amaro et al reviewed 172 patients in

the acute phase of ischemic stroke who received thrombolysis. Specifically, 139 of

them received antiplatelet aggregation therapy within 24 hours, 33 received

corresponding therapy after 24 hours as usual. The results showed that the vascular

recanalization rate of the early antiplatelet aggregation treatment group was higher at

3 days, the neurological function of the patients in the early antiplatelet aggregation

treatment group was significantly better than that of the conventional treatment group

at 90 days, and there was no significant difference in sICH ratio between the two

groups [107]. In another multicentre retrospective study in Europe, 3,782 (31.9%) of

the 11,865 stroke patients who received intravenous thrombolysis were routinely

treated with antiplatelet drugs, mostly aspirin (3,016 patients, accounting for 25.4%),

The results showed that the incidence of sICH and short-term mortality in patients

receiving antiplatelet aggregation therapy during thrombolysis were not significantly

different from those who did not receive antiplatelet aggregation therapy during

thrombolysis [108]. Therefore, for thrombolytic patients, early use of aspirin may

improve the prognosis without increasing the probability of sICH.

A multi-centre, randomized, open study from Germany started intravenous aspirin

therapy early 90min after intravenous thrombolysis compared with intravenous rt-PA

alone. All stroke patients in the experimental group were treated with oral aspirin 24 h

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 43

after intravenous rt-PA. It was planned to include 800 patients. After 642 patients

were studied, the trial was finished ahead of schedule because the incidence of

symptomatic intracranial haemorrhage in the aspirin treatment group was extremely

high. The result analysis showed that the possibility of symptomatic intracranial

haemorrhage in the intravenous aspirin combined with rt-PA group was twice as high

as that in the intravenous rt-PA group alone, and there was no significant difference in

the 90-day outcome between the two groups (mRS score 0-2 points).

(II)

Clopidogrel is a prodrug. Active metabolites of clopidogrel irreversibly bind to ADP

receptors on the surface of platelets, thus inhibiting platelet aggregation. A

randomized controlled trial (CAPRIE) has confirmed that the incidence of vascular

events (ischemic stroke, myocardial infarction or vascular death) after clopidogrel

75mg/d treatment was significantly lower than aspirin 75mg/d treatment [109].

Compared with aspirin, clopidogrel has obvious advantages in symptomatic

atherosclerotic patients with a higher risk of recurrence of cardiovascular and

cerebrovascular diseases within 3 years of follow-up [110]. A meta-analysis shows that

clopidogrel reduces the risk of stroke recurrence by 32% compared with placebo, but

the recurrence rate still reaches 8.5% [111], The reason may be related to clopidogrel

resistance, and its incidence rate is as high as 31% [112]. The carrying rate of CYP2C19

intermediate metabolism and slow metabolism in Asian population is much higher

than that in European and American population.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 44

A single oral dose of 300-600 mg clopidogrel can rapidly inhibit platelet aggregation.

A preliminary prospective study included 20 patients with an average onset time of

25h after stroke, who were given clopidogrel 600mg orally. No deterioration of

neurological function or intracranial haemorrhage was reported [113].

(III)

Ticagrelor is an , whose mechanism of action is reversible binding

with P2Y12 receptor on the surface of platelet membrane, inhibiting ADP-mediated

platelet aggregation, and is not affected by CYP2C19 genotype. In the

ONSET/OFFSET study that compared the antiplatelet effect of clopidogrel, ticagrelor

took effect faster and had stronger effect, and platelet function recovered faster after

withdrawal of the drug [114]. In the RCT named PLATO, the analysis results of the

stroke /TIA subgroup (n=1152) showed that ticagrelor had a tendency to reduce the

primary outcome events and mortality compared with clopidogrel [115].The 2016

SOCRATES study showed that there was no significant difference in the incidence of

major endpoint events (including haemorrhagic stroke or ischemic stroke, myocardial

infarction and death) between ticagrelor and aspirin for AIS or TIA patients (P=0.07).

However, in the secondary endpoint analysis, ticagrelor can significantly reduce the

recurrence of ischemic stroke by 13%(P=0.046) [116]. The subgroup analysis results of

the SOCRATES study shows that among Asian patients with TIA and minor stroke,

the risk of stroke recurrence, myocardial infarction or death is lower in the ticagrelor

group (10.6% vs. 5.7%, P< 0.01), and there is no difference in major bleeding risk

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 45

between the two groups (0.6% vs. 0.8%, HR=0.76, 95% CI: 0.36-1.61). The subgroup

analysis results of another SOCRATES study show that patients with AIS or TIA

accompanied by ipsilateral intracranial artery stenosis are at a lower risk of stroke

recurrence, myocardial infarction or death than the aspirin group (HR=0.68, 95% CI:

0.53-0.88, P=0.003) [117].

(IV)

Cilostazol can reversibly inhibit platelet aggregation and antithrombotic formation,

inhibit vascular smooth muscle cell proliferation and protect vascular endothelial cells

by inhibiting the activity of cyclic phosphodiesterase-3 (PDE-3) and

inhibiting the degradation of cyclic adenosine monophosphate (cAMP).

Huang Yining et al [118] conducted a randomized, double-blind prospective study

(CASISP), where 720 patients were randomly divided into two groups, treated with

aspirin and cilostazol respectively. The results show that both drugs can reduce the

recurrence rate of stroke without obvious difference, but the incidence rate of

haemorrhagic complications in the cilostazol group is significantly lower, suggesting

cilostazol is more suitable for cerebral infarction patients with higher risk of

haemorrhage. In the CSCP Phase-II study, 2,672 patients were enrolled in 278

hospitals in Japan, including 1337 patients in the cilostazol group and 1335 patients in

the aspirin group. The number of patients requiring hospitalization due to various

stroke events in the cilostazol group was significantly lower than that in the aspirin

group. The study believes that cilostazol is no worse than aspirin in secondary

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 46

prevention of ischemic stroke, and has fewer bleeding events, and can be taken on a

long-term basis [119].

(V)

Indobufen exerts antiplatelet effect by reversibly inhibiting COX-1 and inhibiting

platelet aggregation induced by ADP, epinephrine, collagen and arachidonic acid [120].

In addition, it can also reduce platelet factor 3 and platelet factor 4, significantly

inhibit coagulation factor II and coagulation , and has anticoagulant effect

[121].

A study conducted a one-year follow-up of 270 patients with TIA who took indobufen

(100mg/ time, bid), and the results showed that the incidence of TIA was significantly

reduced after one-month treatment (P< 0.001) [122]. In addition, a domestic study

randomly divided 170 patients with acute cerebral infarction into the indobufen group

and aspirin group to compare clinical effects. The results showed that the neurological

impairment scores of the two groups were significantly improved, and there was no

statistical difference. The incidence of adverse reactions possibly related to taking of

indobufen and aspirin were 2.4% and 5.7% respectively, and indobufen was relatively

safe to use during acute cerebral infarction [123].

The post-marketing monitoring study of indobufen included 5,642 patients with

atherosclerotic ischemic vascular disease, 40% of whom took 200mg of indobufen

daily, 60% of whom took 400mg of indobufen daily, 76% of whom took continuous

treatment for at least 3 months. During the study, 220 patients (3.9%) had adverse

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 47

reactions. Specifically, the incidence of gastrointestinal adverse reactions was 3.8%,

and the incidence of haemorrhage-related events was only 0.38% [124].

A meta-analysis involving 19 RCTs with a total of 5,304 patients found that 7 RCTs

reported the incidence of stroke. The analysis results showed that indobufen was not

statistically significant from aspirin and warfarin in the incidence of stroke. Compared

with aspirin, indobufen has a lower incidence than the aspirin group in terms of

haemorrhage, gastrointestinal reaction and total adverse drug events [125].

(VI) Abciximab

In 2000, neurological researchers at the University of Iowa conducted a prospective,

multi-center, double-blind, placebo-controlled study on the safety (24-hour

symptomatic/asymptomatic cerebral haemorrhage) and efficacy (3-month mRS score,

Barthel index) of abciximab [126]. The study found that the safety of abciximab was

similar to placebo and did not significantly improve the 3-month functional prognosis.

The ABeSTT trial was a global, double-blind, placebo-controlled RCT. After 808 AIS

patients were enrolled, it was terminated earlier due to the unsatisfactory benefit-risk

ratio of abciximab. Intravenous administration of abciximab did not improve the

functional prognosis of patients, and increased the risk of intracranial haemorrhage

[127].

There was a trial in 2002 and 2010 respectively that evaluated the safety and efficacy

of intravenous thrombolysis combined with abciximab for AIS patients. Lee et al[128]

found in a small sample pre-study in 2002 that intravenous thrombolysis combined

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 48

with abciximab can significantly improve the functional prognosis of patients without

increasing the risk of bleeding. This conclusion needs further confirmation in large-

scale prospective studies. In 2010, Gahn et al [78] conducted a trial of reduced

thrombolysis combined with abciximab in the treatment of AIS and reached a similar

conclusion. The sample size of the above two studies was less than 30 patients,

resulting in low referability. There is no prospective, multicenter RCT on the

secondary prevention and treatment effect of abciximab on cerebral infarction in

China.

(VII)

Tirofiban is a platelet surface glycoprotein Ⅱb/Ⅲ receptor antagonist. Kellert et

al[129]found that the risk of adverse prognosis and bleeding of bridging tirofiban was

increased compared with thrombectomy alone in a prospective cohort study released

in 2013 on bridging tirofiban for antithrombotic treatment after mechanical

thrombectomy.

Lin et al [130] In the antithrombotic study on AIS patients beyond thrombolytic time

window released in 2017, tirofiban was found to improve functional prognosis

without increasing bleeding risk compared with aspirin or clopidogrel. The results of

the 25-sample study need to be further verified in large-sample prospective trials. The

SaTIS trial was a prospective, multicenter, placebo-controlled, open-label RCT with a

total of 260 samples. The study found that tirofiban did not significantly improve the

functional prognosis compared with placebo, but did not increase the risk of bleeding

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 49

and could reduce mortality [131].

In recent years, the research on tirofiban bridging thrombectomy/thrombolysis,

combined with anticoagulant drugs/other antithrombotic drugs has a significant

growth trend in China. In 2017, two retrospective cohort studies in China found that

small-dose intravenous tirofiban did not increase the risk of haemorrhage after

mechanical thrombectomy [132]. One of the trials proved that this treatment method

can improve the prognosis of patients [133]. In 2017, Liu Zhiqiang et al [134]carried out

a comparative evaluation trial on the safety and efficacy of antithrombotic therapy for

recurrent TIA. The trial found that tirofiban bridging clopidogrel+aspirin dual

antibody can obtain better curative effect and reduce adverse reactions compared with

clopidogrel+aspirin dual antibody alone, which is more worthy of promotion. In 2017,

Wang Sheng et al [135]carried out a trial on antithrombotic therapy for progressive

stroke (tirofiban bridging clopidogrel+aspirin dual antibody, compared with

clopidogrel+aspirin dual antibody alone), and the researchers also reached similar

conclusions.

(VIII)

Eptifibatide selectively blocks the binding of adhesion protein to glycoprotein Ⅱb/Ⅲa.

The CLEAR study is a prospective, multi-center, double-blind RCT. The study found

that there is no significant difference in safety and efficacy between low-dose

intravenous thrombolytic bridging eptifibatide and standard thrombolytic therapy, and

the standard-dose thrombolytic therapy shows a trend of better efficacy.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 50

The CLEAR researchers conducted a post hoc analysis and propensity matched

analysis on a series of studies conducted in 2008, 2013 and 2014 [136]. It found that

low-dose intravenous thrombolysis bridging eptifibatide is not superior to standard

thrombolysis in improving the 90d mRS score, and further confirmation by

prospective RCT is needed. In 2015, the CLEAR researchers conducted a single

cohort, open-label multi-center trial [137]. The trial found that the safety of standard-

dose thrombolytic bridging eptifibatide seems to need further confirmation by

prospective RCT. There is still a lack of prospective, multicenter RCT of eptifibatide

for secondary prevention of cerebral infarction in China.

Recommendations

[1] Aspirin is recommended for patients with AIS within 24 to 48 hours after

onset. For patients treated with IV rt-PA, aspirin is usually delayed until 24

hours later (Class I, Level of Evidence A).

[2] Aspirin (50-325 mg/d) or clopidogrel (75 mg/d) alone can be used as primary

antiplatelet drug therapy (Class I, Level of Evidence A).

[3] Aspirin therapy is not recommended as an alternative therapy for AIS

patients who are suitable for rt-PA IV thrombolysis or mechanical

thrombectomy (Class III, Level of Evidence B).

[4] Ticagrelor (instead of aspirin) is not recommended for acute mild stroke

(Class III, Level of Evidence B).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 51

[5] Cilostazol can be used in AIS patients as an alternative to aspirin if aspirin or

clopidogrel is not available (Class IIa, Level of Evidence A).

[6] For high risk of ischemic stroke patients with aspirin intolerance

(gastrointestinal adverse reactions or allergies, etc.), indobufen (100 mg per time,

twice a day) is feasible (Class IIb, Level of Evidence B).

[7] Abciximab is not recommended for AIS (Class III, Level of Evidence B).

[8] Tirofiban is safe in the perioperative period for bridging therapy or

endovascular therapy. The recommended dose is 0.1-0.2 ug/(kg•min) and

continuous infusion should be limited in 24 hours (Class IIa, Level of Evidence

B).

[9] The efficacy of tirofiban and eptifibatide has not been fully determined, and

further studies are needed to confirm (Class IIb, Level of Evidence B).

II. Dual antiplatelet therapy

(I) Clopidogrel combined with aspirin

The study of Fast Assessment of Stroke and TIA to Prevent Early Recurrence

(FASTER) randomly grouped the TIA or mild ischemic stroke patients within 24

hours of onset, and gave aspirin combined with clopidogrel dual antiplatelet therapy

and aspirin monotherapy to them to observe and compare the 90d clinical prognosis

of the patients. The results showed that the 90-day stroke risk of patients receiving

combined therapy was 7.1%, compared with 10.8% in the monotherapy group. Early

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 52

dual antiplatelet therapy reduced the absolute risk of stroke recurrence and did not

increase the risk of intracranial haemorrhage. The trial was stopped because the case

collection was too slow and no definite conclusion could be reached [138]. The study of

the Management of Atherothrombosis with Clopidogrel in High-Risk Patients with

Recent Transient Ischemic Attack or Ischemic Stroke (MATCH) The study showed

that in patients with recent ischemic stroke or high-risk TIA, after a follow-up of 3.5

years, it was found that the primary outcome events (including ischemic stroke,

myocardial infarction, vascular death and readmission due to acute ischemic events)

of aspirin combined with clopidogrel treatment group had no significant difference

from the clopidogrel monotherapy group (P=0.224), but increased the risk of massive

haemorrhage and fatal haemorrhage [139]. In the secondary prevention (SPS3) study of

AIS subcortical minor stroke, there was no significant difference in the recurrence

rate of ischemic stroke disability or fatal stroke between the clopidogrel combined

with aspirin (325mg/d) group and aspirin monotherapy group, but the haemorrhage

and death risks of the combined therapy group were significantly increased [140]. Dual

antiplatelet therapy was not recommended in the global guidelines before 2011 for

patients with ischemic stroke. The release of the 2013 study of Clopidogrel and

Aspirin versus Aspirin Alone for the Treatment of High—risk Patients with Acute

Non-disabling Cerebrovascular Event (CHANCE) has changed this status.

The CHANCE study included 5170 patients with acute mild ischemic stroke or TIA

with high recurrence risk, and compared the efficacy and safety of clopidogrel

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 53

combined with aspirin dual antiplatelet therapy and aspirin monotherapy. The patients

in the trial group were treated with clopidogrel for 3 months (the loading dose on the

first day was 300mg, then 75mg/d) and aspirin (75mg/d) was used 21 days before the

combination, while the patients in the control group were treated with aspirin

(75mg/d) alone. The results showed that compared with aspirin alone, the dual

antiplatelet therapy group reduced the relative risk of 90 days stroke by 32%, reduced

the absolute risk by 3.5%, and did not increase the haemorrhage risk. The CHANCE

study confirmed that compared with aspirin monotherapy, clopidogrel combined with

aspirin can significantly reduce the 90-day stroke recurrence risk without increasing

the incidence of moderate and severe haemorrhage in high-risk patients with acute

non-disabling cerebrovascular events [141]. Meta-analysis data show that short-term

aspirin combined with clopidogrel is more effective than monotherapy and does not

increase the risk of haemorrhage [142]. Two randomized controlled trials published in

2014 and 2015 both confirmed that aspirin combined with clopidogrel antiplatelet in

early AIS was superior to aspirin alone in reducing the deterioration and recurrence of

the nervous system in the early phase [143]. The POINT study published in May 2018

showed that compared with aspirin alone, clopidogrel combined with aspirin dual

antiplatelet therapy can reduce the risk of severe ischemic events in patients with mild

ischemic stroke or high-risk TIA [162]. The results of the CHANCE study have been

confirmed in European and American population. All the above studies have

confirmed that aspirin combined with clopidogrel in the early phase is superior to

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 54

aspirin alone in reducing the recurrence of stroke and improving the prognosis of

neurological function.

(II) combined with aspirin

In 1999, Millan-Guerrero et al [145]found that comparing dipyridamole intravenously

given to AIS patients 24 hours after onset with oral aspirin, aspirin was more

advantageous in improving functional prognosis. Haungsaithong et al[146] in a small-

sample RCT, it was found that aspirin combined with dipyridamole antiplatelet

therapy had no advantage in AIS treatment compared with aspirin, clopidogrel and

cilostazol antithrombotic monotherapy. In contrast, clopidogrel could significantly

improve the NIHSS score of stroke patients in the 4th week after onset.

In 2005 and 2010, Chairangsarit and Dengler et al compared the therapeutic effects of

aspirin combined with dipyridamole dual antithrombotic therapy and aspirin

respectively. Chairangsarit[147] in a pre-test with a sample size of 38, it was found that

aspirin combined with dipyridamole in 48 hours after onset could significantly

improve neurological impairment symptoms 6 months after onset of the disease than

aspirin antithrombotic therapy, but there was no significant difference in reducing

recurrence rate between the two. Dengler et al[148] in a prospective, multi-center and

large-sample study, found that after aspirin and dipyridamole were given to AIS or

TIA patients within 24 hours of onset, there was no significant difference in

improvement of 90d mRS score, vascular events, etc. compared with aspirin

monoclonal antibody first given for 7 days and then changed to dual antithrombotic

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 55

therapy. In the subgroup analysis of the PRoFESS research published in 2010, Bath et

al [149] found that aspirin combined with sustained-release dipyridamole had similar

effects in improving functional prognosis, recurrence rate and mortality rate, and had

no statistical difference in safety compared with clopidogrel.

There is still a lack of prospective, multicenter RCTs of dipyridamole for secondary

prevention of cerebral infarction in China. Liang Tao et al [150] found that aspirin

combined with dipyridamole was more effective and safe in preventing cerebral

infarction than aspirin alone in a case-control trial with a sample size of 178 in 2010.

Recommendations

[1] For patients with mild stroke and high-risk TIA who did not receive IV

thrombolysis, dual antiplatelet therapy [aspirin 100 mg/d, clopidogrel 75 mg/d

(first day load dose 300 mg)] was initiated within 24 hours of onset and lasted for

21 days, then clopidogrel 75 mg/d which could significantly reduce stroke

recurrence for 90 days (Class I, Level of Evidence A).

[2] The efficacy of dipyridamole alone or dipyridamole combined with aspirin of

preventing the recurrence of ischemic stroke still needs RCTs to confirm (Class

IIb, Level of Evidence B).

III. Triple antiplatelet therapy

The TARDIS study released in 2017 compared the effects of triple antibody (aspirin,

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 56

clopidogrel and dipyridamole) and monoclonal antibody (clopidogrel) or double

antibody (aspirin combined with dipyridamole) treatment on the recurrence rate and

severity of stroke and TIA within 90 days. Specifically, the treatment time of triple

antibody was 30 days, and the antiplatelet treatment schemes of the two groups were

the same after 30 days. The subjects of the study were acute non-cardiogenic ischemic

stroke and TIA patients over 50 years old, who were randomly assigned to the

experimental group and control group. The median time of randomization was 29.3h.

The study plannned to include 4,100 patients and was stopped after 3,096 patients

were included. The reason was that although there was no statistical difference in the

recurrence rate and severity within 90 days ( 95%CI: 0.67-1.20, P=0.47) between the

triple treatment group and the single or dual treatment group, the incidence rate of

bleeding complications in the triple treatment group was 20%, and the incidence rate

of bleeding complications in the monoclonal antibody/double antibody therapy group

was 9%, with statistical difference (P< 0.001, 95%CI: 1.25-3.96) [151,152].

Considering that 11% of the patients in the TARDIS trial had a NIHSS score of > 6,

and 10% of the patients had received thrombolytic therapy before randomization,

which may affect the results. In addition, in the TARDIS subgroup analysis, patients

with mild stroke had higher benefits (P=0.09). Whether limiting the inclusion criteria

to minor stroke and giving triple antibody therapy is more meaningful than

monoclonal antibody/double antibody therapy still need further trials and discussions.

The randomization time of 70% patients in the TARDIS trial was 24-48h after onset.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 57

Further attention should be paid to whether the delay in starting the triple antibody

time window will affect the randomization time, and to the subgroup analysis within

24h after onset and 24h after onset. In addition, the CHANCE, SOCRATES and

TIAregistry.org suggest that most patients relapse within 8-10 days after onset. The

TARDIS study limits the duration of triple antibody to 30 days to be too long, and

adjusts it to 8-10 days to reduce the incidence of bleeding complications. Further

clinical research is needed.

Recommendations

Triple antiplatelet aggregation therapy (aspirin, clopidogrel and dipyridamole)

are not recommended for the treatment of acute non-cardiogenic stroke and TIA

(Class III, Level of Evidence B).

See Supplemental Table 8 for a summary of commonly used antiplatelet drugs for

acute ischemic stroke.

Please refer to Figure 4 for details of the anti-platelet aggregation treatment process

for patients with acute ischemic stroke.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 58

Section 5 Other treatment in acute phase

I. Neurovascular protection

Several studies have shown that butylphthalide could improve collateral circulation

and rescue the penumbra [153-155]. The results of multi-centre randomized controlled

trials showed that butylphthalide was safe and effective in the treatment of acute

cerebral infarction, and could improve neurological impairment and daily living

ability. A meta-analysis involving 21 trials (2123 patients) has also confirmed that

butylphthalide could effectively improve neurological impairment in AIS patients

with few or no serious adverse reactions [156].

In a multicentre, randomized, double-blind, placebo-controlled trial that evaluated

intravenous use of human urinary kallidinogenase (Urinary Kallidinogenase) in

patients with acute cerebral infarction, the human urinary kallidinogenase treatment

group showed greater improvements in functional outcomes and was safer than the

placebo group [157].

Vinpocetine can selectively expand cerebral vessels by inhibiting phosphodiesterase

type I enzyme, reduce cerebral vascular resistance, increase cerebral blood flow

perfusion in focal areas, increase consumption and utilization of glucose and oxygen

in brain tissues without affecting systemic circulation (blood pressure, cardiac output,

heart rate and total peripheral resistance), thus reduce neurological impairment, and

has been widely used to treat cerebrovascular diseases. In terms of neuroprotection,

many studies have found that vinpocetine can inhibit the release of inflammatory

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 59

factors and the activation of microglia by inhibiting the NF-κB signaling pathway,

thus playing a neuroprotective role. A multicenter, randomized controlled, evaluator-

blind study on ischemic stroke patients with onset time of 4.5~48h showed that

vinpocetine can inhibit the inflammatory response level in the area around the

infarction and the inflammatory factor level in the peripheral blood, and can inhibit

the expansion of the infarction volume, thereby improving clinical prognosis,

relieving the cytotoxic effect induced by excitatory amino acids, inhibiting sodium

iron and calcium ion channels, and enhancing the neuroprotective effect of adenosine.

It interferes with atherosclerosis by reducing proliferation and migration of vascular

smooth muscle, inhibiting rational remodeling of vascular diseases [158-160].

Edaravone is a free radical scavenger and antioxidant. Pre-clinical and clinical studies

have shown that edaravone treatment can reduce the incidence of complications such

as haemorrhagic transformation, progressive stroke, epilepsy, pulmonary infection

and so on in patients with diabetic ischemic stroke [161-163].The subgroup analysis of

the RESCUE-Japan Registry study found that edaravone combined with intravenous

rt-PA or intravascular treatment group achieved a good prognosis proportion of

neurological function in AIS patients with macroangiopathy within the treatment time

window [164].

In a small preliminary rial, AIS patients were well tolerated with erythropoietin,

which may improve the functional outcome for one month and reduce the occurrence

of post-stroke complications. However, it has not been proved by other trials, and the

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 60

data failed to show an improvement in prognosis [165-167].

Ginkgolides mainly inhibit platelet aggregation and promote the recovery of nerve

function by antagonizing the platelet activating factor (PAF) [168].A multicentre,

randomized, double-blind, placebo-controlled clinical study of 949 patients with

ischemic stroke within 72 hours is also underway to observe the effect of AIS

combined with Ginkgolide Injection on neurological outcome [169].

Prostaglandin E1 (Alprostadil, PGE1) plays an important role in the treatment of

ischemic stroke by reducing the generation of oxygen free radicals and the release of

proteolytic enzymes, reducing the synthesis and release of TXA2, and protecting

vascular endothelium [170].

A randomized controlled study on showed that it was safe and effective for

AIS patients [171].Several studies of N-methyl-D- receptor glycine site

antagonist suggest good tolerance, but the application in AIS patient within 6h fails to

improve prognosis [172-174].

Preliminary trials on showed that lubeluzole is safe [175], and can reduce

mortality, but the following two larger-scale clinical trials found that lubeluzole is

ineffective in reducing post-stroke mortality and improving prognosis [176-177]. NXY-

059 is a free radical scavenger, and preliminary trials have showed good tolerance

[178]. A study showed that it may reduce the disability rate and intracranial

haemorrhage [179]. However, a key study to confirm the efficacy has not found any

benefits [180].

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 61

Magnesium is an excitatory amino acid antagonist, calcium channel blocker and

cerebral vasodilator. Although preliminary studies show that has good

tolerance [181], and may improve prognosis [182], the results of subsequent larger-scale

clinical trials are negative [183]. Several small-scale studies in China suggest that

magnesium sulfate is effective for early treatment [184,185]. Considering the effect of

application time on curative effect, a study preliminarily confirmed the safety and

feasibility of pre-hospital ultra-early magnesium sulfate treatment [186]. A large-scale

clinical trial showed that it is safe to start magnesium sulfate treatment before

admission, and treatment is allowed to start within 2 hours after stroke occurs,

however, it does not improve the 90-day disability outcome [187].

Several clinical trials on the efficacy of citicoline have failed to prove its efficacy on

AIS [188-190]. However, a meta-analysis on the research level showed that citicoline has

a net benefit in reducing the disability rate [191].The meta-analysis shows that if drug

therapy is started within 24 hours after symptoms occur, patients with moderate to

severe stroke may benefit [192].

Several trials on ganglioside (GM1) have shown that it cannot improve the prognosis

[193,194]. A systematic review of these drugs [195] also failed to show benefits in

treatment. However, a small-scale study in China have shown that

combined with gangliosides could improve the short-term neurological deficit and 90

days activity of daily living in AIS patients (superior to the aspirin monotherapy or

argatroban sequential aspirin treatment group) [196].

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 62

II. Hypothermia

Experimental and local hypoxic brain injury models suggest that hypothermia has

neuroprotective effects. Deep hypothermia is often used to protect brain tissue in

major operations.

An RCT was performed to compare the mortality and neurological prognosis of

patients with cerebral hemisphere infarction between the mild hypothermia group and

normal body temperature group, there was no statistical difference in mortality, but

the mild hypothermia had a better neurological prognosis [197]. A study evaluating

mild hypothermia therapy with surface cooling device after intravenous thrombolysis

showed that mild hypothermia therapy was relatively safe and feasible despite some

adverse events compared with normal body temperature [198]. The results of two

studies on intravascular hypothermia therapy for patients after intravenous

thrombolysis showed that hypothermia therapy was safe and feasible, but it had no

obvious clinical benefits and can not reduce the incidence of pneumonia [199, 200]. The

ReCCLAIMI Phase I study mainly evaluated the feasibility and safety of intravascular

hypothermia immediately after arterial reperfusion therapy. Compared with patients

with historical control, it is proved that hypothermia can prevent intracerebral

haemorrhage (OR=0.09, 95% CI: 0.02-0.56, P< 0.01), and is safe and feasible [201]. A

prospective cohort study compared the safety and prognosis of the mild hypothermia

group and control group in AIS patients after successful revascularization. The results

showed that the mild hypothermia group (n=39) had less cerebral oedema (P=0.001),

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 63

haemorrhagic transformation (P=0.016) and better outcomes (P=0.017) than the

normothermia group (n=36). Conclusion: For patients with ischemic stroke, after

successful recanalization, hypothermia therapy may reduce the risk of cerebral

oedema and haemorrhagic transformation, and lead to improvement of clinical

outcomes [202].

With regard to different cooling methods, a randomized trial compared the safety and

cooling ability of intravascular cooling and surface cooling, showing that hypothermia

therapy is feasible for patients under general anesthesia, the incidence of pneumonia

in the hypothermia treatment group increases, and intravascular cooling has a faster

induction period than surface cooling [203].

As far as cooling temperature is concerned, a multi-center RCT evaluated the

feasibility and safety of surface cooling at different target temperatures in conscious

patients with AIS. The conclusion is that in the conscious AIS patients, the surface

cooling could reach 35.0℃, but could not reach 34.5℃ and 34.0℃, cooling was

associated with increased risk of pneumonia. However, the study was terminated after

22 subjects were enrolled due to slow recruitment [204]. Two observational studies

evaluated the safety and feasibility of moderate hypothermia and its potential to

reduce intracranial pressure, and concluded that moderate hypothermia is feasible in

patients with acute stroke, although it is related to several side effects. Most deaths

occur during rewarming due to rebound of intracranial pressure [205, 206].

III. Hyperbaric oxygen

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 64

Although preclinical studies on acute cerebral ischemia show that hyperbaric oxygen

therapy is usually relatively beneficial, in clinical trials, the efficacy of hyperbaric

oxygen therapy for AIS is uncertain or shows that this measure does not improve the

prognosis. A small sample RCT compared the functional outcomes of the hyperbaric

oxygen treatment group and the control group in the early phase (2 weeks after onset)

and the late phase (1 month after onset), the results showed that there was no

significant difference in the early curative effect, but there was significant difference

in the late curative effect, indicating that hyperbaric oxygen treatment may be

effective for AIS patients [207]. Many small sample studies in China [208-209] and two

meta analyses [210-211] have shown that hyperbaric oxygen or hyperbaric oxygen

combined with drug therapy is beneficial to AIS patients, but due to its small sample

size and relatively poor research quality, its conclusion cannot be used as effective

evidence. In a review of 11 RCTs involving 705 AIS participants treated with

hyperbaric oxygen, the results showed that there was no difference in 6-month

mortality between the HBO treatment group and the control group (RR=0.97, 95%

CI: 0.34-2.75, P=0.96). However, 4 of the 14 disability and functional outcome

indicators showed that hyperbaric oxygen treatment had certain benefits. The

conclusion is that there is no evidence that hyperbaric oxygen treatment can improve

AIS outcomes, although clinical benefits cannot be excluded. In addition, due to

different test methods, it is difficult to summarize and analyse the results other than

the mortality rate [212]. Therefore, current research data do not support routine

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 65

applications of hyperbaric oxygen treatment for AIS.

Regarding hyperbaric oxygen treatment, most studies have proved that it is safe, but

there are still some side effects. A review has mentioned that middle ear barotrauma is

the most common complication, and other side effects include paranasal

sinus/paranasal sinus barotrauma, dental barotrauma, pulmonary barotrauma,

increased blood pressure, claustrophobia, and epileptic seizures [213]. A retrospective

study involved 931 patients and underwent 23,328 times of hyperbaric oxygen

therapy, mainly assessing the overall incidence of epilepsy and the risks under

different treatment pressures. The results showed that the seizure rate was 1 seizure in

2121 treatments (about 5 seizures per 10000 treatments), and the higher the pressure,

the more frequent the seizure. The conclusion is that hyperbaric oxygen treatment is

related to the increased risk of epileptic seizure. The higher the pressure, the greater

the risk. However, the study population was all patients treated with hyperbaric

oxygen, not only the ischemic stroke patients [214].

Hyperbaric oxygen treatment is a recognized treatment method for cerebral air

embolism related to compressed-air diving accidents, thus it is also the standard

treatment for iatrogenic causes of gas embolism. A study retrospectively analysed the

characteristics of 36 patients with cerebral gas embolism (CAGE) and the prognosis

of hyperbaric oxygen treatment, which showed that a high proportion of CAGE

patients who received hyperbaric oxygen treatment had good curative effect. The time

from onset to hyperbaric oxygen treatment ≤ 6h can increase the possibility of

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 66

favourable outcome, while CT/MRI scan before hyperbaric oxygen treatment found

that infarction/oedema had reduced the possibility of favourable outcome [215]. Stroke

is a rare but serious complication in cardiac surgery. In most cases, cerebral gas

embolism is the possible cause. A study on AIS hyperbaric oxygen therapy after

cardiac surgery shows that hyperbaric oxygen treatment is related to significant

improvement of acute neurological impairment caused by ischemic stroke after

cardiac surgery. Although gas embolism is the most likely cause of stroke in this type

of patients, other potential pathological mechanisms cannot be excluded and need

further evaluation through random studies [216].

IV. Mechanical blood flow increase method

The mechanical method to increase cerebral perfusion is a method to improve

cerebral blood flow through the Will circle and pia mater vessels, rather than through

the action of vasoconstrictors. In 1997, a study led by Applebaum et al [217] aimed to

evaluate the effect of external counterpulsation (ECP) on cerebral blood flow and

renal blood flow. The results show that this non-invasive treatment may be helpful to

support patients with cerebral perfusion and renal perfusion reduction. In 2008,

Han[218] also found that ECP is beneficial to patients with macrovascular ischemic

stroke. In recent years, other research results have proved that ECP is safe and

feasible, applicable to AIS, and it is related to the improvement of the NIHSS score

regardless of the pressure used [219]. However, some studies have drawn different

conclusions, and believed that NeuroFlo therapy has nothing to do with the clinical

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 67

outcome of AIS [220]. After that, Hammer et al[221]pointed out that it may be safe and

feasible to use NeuroFlo catheter for partial aortic occlusion within 8-24 hours after

onset of symptoms when evaluating the safety and feasibility of NeuroFlo in patients

with ischemic stroke within 8-24 hours after onset.

Recommendations

[1] Evidence in basic and preclinical studies suggests that neurovascular

protection therapy may be beneficial, but has not been proved in clinical studies

to improve prognosis of AIS patients and reduce recurrence. More clinical

research is still needed, some related researches are also in progress currently

(Class IIb, Level of Evidence B).

[2] The efficacy of induced hypothermia in patients with ischemic stroke is

unclear and further studies are needed. Most studies have found that induced

hypothermia was a risk for infection, including pneumonia. Induced

hypothermia should be administered only in clinical trials (Class IIb, Level of

Evidence B).

[3] Hyperbaric oxygen therapy is not recommended for AIS patients unless it is

caused by air embolism. Hyperbaric oxygen therapy is related to claustrophobia,

middle ear barotrauma and the increased risk of seizures (Class III, Level of

Evidence B).

[4] Mechanical augmentation of blood flow to treat AIS patients has not been

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 68

perfected, curative effect is not sure, and only can be used in clinical trials (Class

IIb, Level of Evidence B).

V. Induced hypertension

Elevated systemic blood pressure in patients with acute ischemic stroke can increase

the local cerebral blood flow, and increase the local cerebral blood flow through

lateral branches and arterioles. Many research results show that induced hypertension

treatment in the acute phase may improve neurological impairment in AIS patients

[222]. However, Koenig et al reviewed 100 AIS patients, of which 46 were treated with

induced hypertension (IH group) and 54 were treated with conventional therapy (ST

group). The results showed that the NIHSS scores of the two groups of patients were

similar in hospital and at discharge, and the recurrence rate of vascular events had no

significant difference between the two groups.

VI. Blood volume expansion and haemodilution

Since the end of 1960s, there has been continuous research on the efficacy of

haemodilution therapy for acute stroke patients, but the research conclusions are

contradictory [223-227]. In 2014, a meta-analysis involving 21 trials and 4,174 samples

showed that there is still no clear evidence to prove that haemodilution therapy is

beneficial to the prognosis of AIS [228]. In 2017, Joseph et al[229] analysed the ALIAS

II data to assess whether the extent of plasma volume expansion affects neurological

function recovery in AIS patients. The results confirmed that plasma volume

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 69

expansion is related to the poor prognosis of neurological function.

VII. Near infrared laser therapy

The randomized controlled study NEST-1 preliminarily confirmed the safety and

efficacy of infrared laser therapy in the treatment of ischemic stroke within 24 hours

after onset [230].The NEST-2 study further confirmed its safety, but its effectiveness

was not significant enough [267]. A summary analysis of the results of NEST-1 and

NEST-2 indicates that there is a statistical difference in the 90d mRS scores, of which

moderate stroke has better response to treatment [231].However, in the NEST-2 study,

the infarct volume of patients was evaluated before and after treatment. The results

showed that laser treatment had nothing to do with the overall or cortical infarct

volume reduction measured by subacute CT [269]. The follow-up Phase III trial was

terminated due to no significant difference in mid-term efficacy analysis. The

conclusion was that transcranial laser therapy had no measurable neuroprotective

effect within 24 hours after AIS onset [234-235].

VIII. Albumin

In many observational studies, albumin reduction is associated with high mortality

and disability in patients with ischemic stroke [236-237]. Relevant studies have

confirmed that albumin therapy has neuroprotective effect in the acute phase [238-240].

The ALIAS study evaluated the efficacy of 6 different doses (0.34-2.05 g/kg) of

albumin in the treatment of AIS. The results showed that the good clinical outcome

(mRS score of 0-1 or NIHSS score of 0-3) rate in the higher dose albumin treatment

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 70

group was 81% higher than that in the lower dose albumin treatment group. The

number of subjects receiving higher dose albumin in the intravenous rt-PA treatment

group who achieved good clinical outcomes was 3 times higher than that in the lower

dose group. Albumin and intravenous rt-PA may have synergistic effect in AIS

treatment [241-242]. However, there are also corresponding research results that confirm

that there is no obvious correlation between the two or that albumin therapy has a

correlation with poor prognosis of patients [243-244]. In 2016, Martin et al. conducted a

joint analysis of the ALIAS Part I and Part II trials. The results showed that there was

no statistical difference in the 90-day disability rate between the two groups [245]. In

2018, Khatri et al analysed 1275 patients in the ALIAS trial to explore the

neuroprotective effect of albumin on AIS patients. The results showed that for patients

with large-area cardiogenic cerebral embolism (NIHSS score ≥ 15), high-dose

albumin therapy started within 2 hours of onset had a trend of obvious benefits,

indicating that certain types of stroke patients might benefit from albumin therapy.

However, further clinical trials are still needed to classify patients according to the

severity of NIHSS, so that treatment can be stratified to further evaluate the clinical

effects of albumin treatment [246].

Recommendations

[1] Effectiveness of induced hypertension treatment in AIS patients is not clear, it

can only be used in the study of clinical research (Class IIb, Level of Evidence B).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 71

[2] It is not recommended for routine use of blood volume expansion or

haemodilution therapy in AIS patients (Class III, Level of Evidence A).

[3] At present, there is no evidence that transcranial near infrared laser therapy

for ischemic stroke is beneficial, therefore using transcranial near infrared laser

treatment in ischemic stroke is not recommended (Class IIb, Level of Evidence

B).

[4] The routine use of high-dose albumin therapy in patients with AIS is not

recommended (Class III, Level of Evidence A).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 72

Section 6 Routine support treatment and complication management for acute

ischemic cerebrovascular disease

I. General supportive treatment

(I) Airway, ventilation support and supplementary oxygen supply

Oxygen deficiency often occurs after stroke [247-248]. The common causes of hypoxia

include incomplete airway obstruction, hypoventilation, aspiration, atelectasis and

pneumonia. Patients with decreased consciousness caused by brainstem dysfunction

increase the risk of airway damage due to oropharyngeal activity damage and loss of

protective reflex [249-250]. A recent RCT suggests that for acute stroke patients without

hypoxemia, preventive low-dose oxygen supply cannot reduce the 3-month mortality

and disability rate [251]. In addition, another cohort study [252] and 5 RCTs[253-257] also

suggest that supplementary oxygen supply to patients with acute stroke has no

definite benefits. According to these data, mild to moderate stroke patients without

hypoxia do not need emergency routine supplementary oxygen supply. Supplementary

oxygen supply for patients with severe stroke may be beneficial, although it has not

been confirmed by the current data. Further research in this field is recommended

[257].According to the data from the review (these data are mainly about patients

recovering from cardiac arrest), the AHA guidelines on acute cardiovascular therapy

for patients recovering from stroke and cardiac arrest recommend oxygen supply to

patients with hypoxemia to maintain a blood oxygen saturation level of > 94% [294].

When there are indications for oxygen therapy, it is reasonable to adopt the least

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 73

invasive method to achieve normal oxygen concentration as much as possible.

Effective methods include nasal catheter, biphasic positive airway pressure

ventilation, continuous positive airway pressure ventilation or endotracheal intubation

mechanical ventilation. At present, there is no clinical trial that evaluates the

effectiveness of tracheal intubation in the management over patients with severe

stroke. It is generally believed that tracheal intubation and mechanical ventilation

should be implemented when the airway is threatened. Evidence shows that early

prevention of aspiration can reduce the occurrence of pneumonia [258], and airway

protection may be an important method for some patients. Tracheal intubation and

mechanical ventilation are also helpful to reduce ICP elevation or malignant brain

oedema after stroke [259-260].Tracheal intubation will affect the prognosis, although a

few patients may obtain satisfactory prognosis [261]. However, the overall prognosis of

stroke patients with tracheal intubation is poor, and the mortality rate within 30 days

after stroke reaches 50% [262-264].

Recommendations

[1] Airway support and ventilator assistance are recommended for the treatment

of patients with acute stroke who have decreased consciousness or who have

bulbar dysfunction that causes compromise of the airway (Class I, Level of

Evidence C).

[2] Supplemental oxygen should be provided to maintain oxygen

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 74

saturation >94%. (Class I, Level of Evidence C).

[3] Supplemental oxygen is not recommended in nonhypoxic AIS patients (Class

III, Level of Evidence B).

(II) Body temperature

About 1/3 of stroke inpatients have fever (body temperature > 37.6℃) in the early

phase after onset [265-266].Fever may be secondary to stroke causes, such as infective

endocarditis, or may be caused by stroke complications, such as pneumonia, urinary

tract infection, UTI), or sepsis. Fever is related to the poor prognosis of neurological

function in AIS patients, and maintaining the normal body temperature or decreasing

the acute elevated body temperature may improve the prognosis of stroke patients

[266].Measures to achieve the normal body temperature or prevent fever include drugs

and physical intervention.

A large-scale randomized, double-blind, placebo-controlled trial evaluated whether

early acetaminophen (acetaminophen) treatment can improve functional prognosis by

lowering body temperature and preventing fever. The results showed that there was no

statistical difference between the two groups. The post-event analysis showed that the

treatment was beneficial to patients with baseline temperature of 37-39 C. The

average hypothermia of patients in the treatment group was 0.26 ℃ (95% CI: 0.18-

0.31) 24 hours after starting the treatment, which was lower than that in the control

group. [267]. Another analysis showed that stroke patients with elevated body

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 75

temperature 24 hours after admission had worse prognosis. The risk of adverse

outcome (OR=1.3, 95% CI: 1.05-1.63) and death (OR=1.51, 95% CI: 1.15-1.98)

increases for every 1 ℃ rise in body temperature [268]. A recent retrospective cohort

study included 9,366 AIS patients from intensive care units in Australia, New Zealand

and the United Kingdom from 2005 to 2013. The results showed that patients with

temperature peaks < 37℃ and > 39℃ within 24 hours after onset had an increased

risk of death during hospitalization compared with patients with normal body

temperature [269].

Hypothermia therapy is a promising neuroprotective strategy, but its benefits to AIS

patients have not been confirmed. Most studies have shown that hypothermia

induction is related to the increased risk of infection including pneumonia [194-195, 204,

270].Hypothermia therapy should only be used in clinical trials.

Recommendations

[1] Sources of hyperthermia (temperature >38°C) should be identified and

treated, and antipyretic should be administered to lower

temperature in hyperthermic patients with stroke (Class I, Level of Evidence C).

[2] The benefit of induced hypothermia for treating patients with ischemic stroke

is not well established. Hypothermia should be offered only in the ongoing

clinical trials (Class IIb, Level of Evidence B).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 76

(III) Nutrition and fluid infusion

Dehydration or malnutrition may delay recovery, so it is very important to maintain

nutrition. Assessment of fluid and nutritional status after stroke should be emphasized,

and fluid infusion and nutritional support should be given when necessary [271-272].

FOOD (Feed or Ordinary Diet; Phase I-III) results show that supplementary diet is

associated with a 0.7% reduction in absolute risk of death, early tube feeding (within

7 days after admission) is associated with a 5.8% reduction in absolute risk of death

and a 1.2% reduction in the incidence of death or poor prognosis. Compared with

percutaneous endoscopic gastrostomy tube feeding, nasal feeding is associated with a

1.0% increase in absolute risk of death and a 7.8% increase in the incidence of death

or poor prognosis [273]. The research conclusion suggests that enteral nutrition should

be started within 7 days after admission of stroke patients. A 2012 Cochrane review

included 6779 patients in 33 RCT studies, and evaluated the efficacy of dysphagia

intervention treatment, feeding strategy and time [early phase (within 7 days) vs. late

phase], fluid infusion and nutritional supplement for patients with acute and subacute

stroke [274]. The conclusion is that there is no obvious difference in the fatality,

mortality and disability rate between percutaneous endoscopic gastrostomy tube

feeding and nasal feeding. Although the data are not enough to determine which of the

two feeding methods are better, percutaneous endoscopic gastrostomy tube feeding

has fewer treatment failures (P=0.007), less gastrointestinal bleeding (P=0.007) and

higher feeding costs (P< 0.00001).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 77

Limited research results show that the implementation of intensive oral hygiene

programs may reduce the risk of aspiration pneumonia. Sorensen et al [275] analysed

the relevant intervention measures for patients with acute stroke. After comparing the

intervention group that adopted standardized dysphagia screening, diet and

standardized chlorhexidine antibacterial oral hygiene program with the historical

control group that did not systematically screen dysphagia within 24 hours and did not

systematically receive chlorhexidine antibacterial oral hygiene program, it was found

that the former standardized intervention measures could reduce the incidence of

pneumonia (7% vs. 28%). Specifically, the efficacy of the standardized oral hygiene

program in the intervention group cannot be separated from standardized dysphagia

screening and diet. In addition, due to the historical nature of the control group, there

may be changes in nursing between patients in the historical control group and the

intervention group, which may affect the risk of occurrence of pneumonia. After a

Cochrane review included three studies, the analysis found that the incidence of

pneumonia in the intervention group of oral care combined with disinfection gel was

lower than that of oral care combined with placebo gel (P=0.03) [276]. Wagner et

al[277]compared the risk of pneumonia before and after systematic oral health care for

stroke inpatients. Compared with the control group, the uncorrected incidence of

hospital-acquired pneumonia in the intervention group with oral health care was lower

(14% vs. 10.33%). P=0.022), and the uncorrected OR was 0.68 (95% CI: 0.48-0.95,

P=0.022). After correction of confounding factors, the OR value of hospital acquired

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 78

pneumonia in the intervention group was still significantly lower, and was 0.71 (95%

CI: 0.51-0.98, P=0.041).

Recommendations

[1] Enteral diet should be started within 7 days of admission after an AIS (Class

I, Level of Evidence B).

[2] For patients with dysphagia, it is reasonable to initially use nasogastric tube

for feeding in the early phase of stroke (starting within the first 7 days) and to

place percutaneous gastrostomy tubes in patients with longer anticipated

persistent inability to swallow safely (>2–3 weeks) (Class IIa, Level of Evidence

C).

[3] Nutritional supplements are reasonable to consider for patients who are

malnourished or at risk of malnourishment (Class IIa, Level of Evidence B).

[4] Implementing oral hygiene protocols to reduce the risk of pneumonia after

stroke may be reasonable (Class IIb, Level of Evidence B).

(IV) Infection prevention

For stroke patients with limited mobility and cough, pneumonia is not only the most

common complication, but also one of the important causes of death after stroke [278-

282]. Aslanyan et al [318] found that pneumonia was associated with increased risk of

death (HR=2.2, 95% CI: 1.5-3.3) or adverse outcomes (OR=3.8, 95% CI: 2.2-6.7).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 79

Stroke-related pneumonia can increase the length of hospitalization, mortality rate and

hospitalization expenses[283]. Limited mobility and atelectasis can lead to pneumonia.

Early activities and good lung care are helpful for preventing pneumonia [284].

Preventive measures for tracheal intubated patients include semi-horizontal

ventilation, airway position, sputum aspiration, early activities, and shortening the

time of intubation when possible [322].Treatment of nausea and vomiting can also

reduce the risk of aspiration pneumonia. Exercise and deep breathing help reduce the

occurrence of atelectasis. For post-stroke fever, physicians should actively search for

signs of pneumonia, and appropriate antibiotic treatment should be given in time.

Some studies have found that early preventive administration of levofloxacin after

stroke has failed to reduce the risk of pneumonia or other infections [285].

Urinary tract infection is common after stroke, which can occur in 15%-60% of

patients. Urinary tract infection is not only an independent predictor of poor

outcomes, but is also is a potential complication that can lead to bacteremia or

septicemia [280, 286-289]. Patients with stroke should have routine urine tests whenever

fever occurs in order to clarify evidence of infection. Some patients have a high risk

of urinary incontinence, especially those with severe disabilities [290]. Catheter

indwelling should be avoided as much as possible, but indwelling may be necessary

in acute stroke, and the catheter should be removed as soon as the patient's condition

is stable. Intermittent catheterization can reduce the risk of infection. External

catheter, paper diaper and intermittent catheter can be used to replace indwelling

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 80

catheter. If a patient's level of consciousness changes and it is determined that there is

no other cause leading to deterioration of neurological function, the presence of

urinary tract infection should be evaluated. If urinary tract infection is suspected,

routine urinalysis and urine culture should be performed [280-281,291]. Urine

acidification can reduce the risk of infection, and anticholinergic drugs may help

restore bladder function. Although preventive antibiotics are not routinely used,

antibiotics should be reasonably applied when the evidence of urinary tract infection

is clear.

Recommendations

[1] Routine use of prophylactic antibiotics has not been shown to be beneficial

(Class III, Level of Evidence B).

[2] Routine placement of indwelling bladder catheters should not be performed

because of the associated risk of catheter-associated urinary tract infections

(Class III, Level of Evidence C).

(V) Prevention of lower limb venous thrombus and pulmonary embolism

CLOTS (Clots in Legs or Stockings After Stroke) 3 is a multi-centre trial involving

2,867 patients in 94 centres in the UK, which compared the preventive effects of

conventional treatment with or without intermittent pneumatic compression (IPC) on

venous thromboembolism in stroke patients with limited mobility. The study included

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 81

acute stroke patients within 3 days of onset and could not independently have bowel

movements. Conventional treatment is defined as possible application of aspirin, fluid

infusion and elastic stockings for non-haemorrhagic stroke. 31% of the patients used

prophylactic, full dose or low molecular heparin, but these patients were evenly

distributed between the two groups. The survival rate of the patients in the IPC group

was significantly improved at 6 months (HR=0.86, 95% CI: 0.73-0.99, P=0.042), but

the disability rate was not improved. Skin damage was more common in the IPC

group (3.1% vs. 1.4%, P=0.002). Contraindications for IPC include local lesions of

lower limbs such as dermatitis, gangrene, severe oedema, venous blood stasis, severe

peripheral vascular disease, postoperative vein ligation, skin transplantation, or

swelling of lower limbs and other signs indicating deep venous thrombosis (DVT)

[292]. A meta-analysis included in the study and 2 other small-scale trials reconfirmed

the above results [293].

In a recent systematic meta-analysis on prophylactic drug intervention of venous

thrombosis in AIS patients included 1 large sample trial (n=14578) and 4 small

sample trials on unfractionated heparin, 8 small sample trials on low molecular

heparin or and 1 trial on heparinoid[293]. Preventive anticoagulants had no

significant effect on mortality and functional status at final follow-up. Symptomatic

pulmonary embolism (OR=0.69, 95% CI: 0.49-0.98) and DVT (mostly

asymptomatic) (OR=0.21, 95% CI: 0.15-0.29) were significantly reduced.

Symptomatic intracranial haemorrhage (OR=1.68, 95% CI: 1.11-2.55) and

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 82

symptomatic extracranial haemorrhage (OR=1.65, 95% CI: 1.0-2.75) increased

significantly. It is possible that the benefits of reduced risk of venous

thromboembolism in some patients are sufficient to offset the increased risk of

intracranial and extracranial haemorrhage, but there is no prediction tool to determine

this part of patients [293-295].

A recent systematic meta-analysis compared the use of low molecular

heparin/heparan and unfractionated heparin to prevent venous thromboembolism in

AIS patients, including 1 large sample trial (n = 1762) and 2 small sample trials

comparing low molecular heparin and unfractionated heparin, and 4 small sample

trials comparing unfractionated heparin and unfractionated heparin. Compared with

unfractionated heparin, low molecular heparin/heparan had no obvious effect on death

or disability. Low molecular heparin/heparan could significantly reduce risk of DVT

(mostly asymptomatic) (OR=0.55, 95% CI: 0.44-0.70), while the risk of severe

extracranial haemorrhage increased (OR=3.79, 95% CI: 1.30-11.03). Low molecular

heparin can be injected once a day, so nursing is more convenient and patients are

more comfortable. It should be noted that for elderly patients with renal impairment,

the use of low molecular heparin may increase the risk of bleeding and increase the

medical expenses.

Recommendations

[1] In immobile stroke patients without contraindications, intermittent

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 83

pneumatic compression (IPC) in addition to routine care (aspirin and hydration)

is recommended over routine care to reduce the risk of deep vein thrombosis

(DVT) (Class I, Level of Evidence B).

[2] The benefit of prophylactic-dose subcutaneous heparin (UFH or LMWH) in

immobile patients with AIS is not well established (Class IIb, Level of Evidence

A).

[3] When prophylactic anticoagulation is used, the benefit of prophylactic-dose

LMWH over prophylactic-dose UFH is uncertain (Class IIb, Level of Evidence

B)

[4] In ischemic stroke, elastic compression stockings should not be used (Class

III, Level of Evidence B).

(VI) Early rehabilitation

When a patient's condition is stable, he or she should start physical activities such as

sitting, standing and walking as soon as possible. When the condition of bedridden

patients permits, attention should be paid to the placement of normal limbs. Attention

should be paid to language, sports, psychology and other aspects of rehabilitation

training in order to restore the ability of daily living as much as possible. The AVERT

(A Very Early Rehabilitation Trial) study randomly assigned 2104 patients with acute

stroke (1: 1) and compared the high-intensity, ultra-early activity programs with the

standard treatment activity programs [296]. However, the findings show that compared

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 84

with the conventional treatment group, the high intensity and ultra-early activity

group had less favourable prognosis (46% vs. 50%), higher mortality (8% vs. 7%),

and more non-lethal serious adverse events (20% vs. 19%). Therefore, the early

activities should be moderate, so as to avoid over early resumption of activities.

Recommendations

[1] It is recommended that early rehabilitation for hospitalized stroke patients be

provided in environments with organized, multidisciplinary stroke care (Class I,

Level of Evidence A)

[2] It is recommended that stroke survivors receive rehabilitation at an intensity

commensurate with anticipated benefit and tolerance (Class I, Level of Evidence

B).

[3] High-dose, very early mobilization within 24 hours of stroke onset should not

be performed because it can reduce the odds of a favourable outcome at 3

months (Class III, Level of Evidence B).

[4] It is recommended that all individuals with stroke be provided a formal

assessment of their activities of daily living and instrumental activities of daily

living, communication abilities, and functional mobility before discharge from

acute care hospitalization and the findings be incorporated into the care

transition and the discharge planning process (Class I, Level of Evidence B).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 85

[5] A functional assessment by a clinician with expertise in rehabilitation is

recommended for patients with an acute stroke with residual functional deficits

(Class I, Level of Evidence C).

II. Management of nervous system complications

(I) Cerebral oedema and signs of mass lesions

1. Medical therapy Cerebral oedema can be seen in all types of cerebral infarction,

especially massive cerebral infarction. Various drug interventions are suggested to

alleviate the development of oedema, such as limiting liquid intake to reduce

hypotonic liquid, avoiding excessive glucose intake, improving hypoxemia and

hypercapnia, and inducing hypothermia therapy. Antihypertensive drugs, especially

those that can cause cerebrovascular contraction, should be avoided. Raising the head

of the bed by 20°-30° is helpful for venous drainage. The goal of these interventions

is to reduce or alleviate oedema formation before intracranial pressure increases

significantly due to cerebral oedema. Standardized intracranial pressure management

should be started when intracranial pressure increases due to cerebral oedema [335].

The intracranial pressure management scheme is similar to treatment for traumatic

brain injury and spontaneous cerebral haemorrhage, including hyperventilation,

hypertonic saline, osmotic diuretics, ventricular drainage of cerebrospinal fluid, and

surgical decompression [297-298]. At present, there is no evidence that hyperventilation,

conventional or high-dose steroid drugs, diuretics, mannitol/glycerol fructose or other

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 86

measures to reduce intracranial pressure can improve the prognosis of patients with

ischemic cerebral oedema. Mannitol (0.25-0.5g/kg) can be given intravenously every

6h for more than 20min to reduce intracranial pressure, and the maximum

conventional dose is 2g/kg. Koenig et al [299]'s preliminary study found that hypertonic

saline can rapidly reduce intracranial pressure in patients with clinical tentorial

herniation caused by various supratentorial lesions (including ischemic and

haemorrhagic stroke), and the results of this stroke-related study supplement and

support the data for previous studies on traumatic brain injury. Hyperventilation is a

very effective treatment for rapidly improving cerebral oedema, but it takes effect by

inducing cerebral vasoconstriction. If persistent hypocapnia occurs, it can aggravate

ischemia [300]. Therefore, hyperventilation should be induced as soon as possible to

avoid excessive hypocapnia (< 30mmHg). At present, the evidence for the treatment

of AIS with hypothermia or is still limited. The existing data only come

from studies of small sample size and unclear intervention time. A recent meta-

analysis involving 6 RCTs showed that hypothermia had no effect on stroke outcome

[301]. Further related studies are recommended. Although active drug management has

been given, the mortality rate of patients with intracranial hypertension is still as high

as 50%-70%. Therefore, these interventions should be considered as temporary

measures to extend the time window before final treatment.

2. Surgical treatment Cerebral hemispheric infarction is often caused by occlusion of

proximal large vessels and is associated with massive cerebral infarction that often

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 87

involves upper and lower brain tissues of lateral fissure [302-304]. Imaging studies

showed that in early CT, low density lesions in more than 2/3 of the middle cerebral

artery area [302], perfusion restriction [305-306] or lack of perfusion [307], all indicate that

the risk of delayed cerebral hernia is increased. Clinical deterioration often progresses

rapidly. When accompanied with brainstem compression, the rapid dysfunction of the

upper part of the brainstem may first lead to deterioration of the consciousness level

[308-309]. Even if the maximum degree of drug administration had been given, the death

rate of patients with consciousness deterioration is still 50%-70% [310]. Brain stem

compression is often accompanied by secondary effects of frontal lobe and occipital

lobe, which may be caused by dural structure compression by anterior and posterior

cerebral arteries [311-312]. The secondary infarction caused by this greatly limits the

potential possibility of clinical rehabilitation and even survival.

Cerebral oedema in patients with supratentorial massive cerebral infarction can lead

to serious and even life-threatening complications. Although less severe brain oedema

can be managed by drug treatment, surgical treatment may be the only effective

choice for patients with severe brain oedema. In this case, timely decompressive

craniectomy can reduce the mortality rate [313]. Summary results of several RCTs

showed that decompressive craniectomy can significantly reduce mortality within 48

hours after onset of malignant middle cerebral artery infarction in patients under 60

years old, and the absolute risk of mortality at 12 months can be reduced by 50%

(95% CI: 34-66) [313]. The study II of "Decompressive Surgery for the Treatment of

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 88

Malignant Infarction of the Middle Cerebral Artery" (DESTINY) showed that

decompressive craniectomy can reduce the mortality rate by about 50% (76% in non-

operative group and 42% in operative group) within 48 hours after stroke onset for

patients with malignant infarction of middle cerebral artery who are aged > 60 years

[314-320]. However, the functional outcome of these elderly patients seemed to be worse

than that of patients aged < 60 years. None of the patients in the two groups achieved

self-care within 12 months (mRS score ≤ 2).

Ventricle drainage is recognized as an effective treatment for acute obstructive

hydrocephalus, and often can effectively relieve the symptoms of patients with acute

ischemic stroke [321-322]. Therefore, ventricular drainage is the reasonable first choice

for patients with cerebellar ischemic stroke with obstructive hydrocephalus. If

drainage of cerebrospinal fluid through ventricle drainage fails to improve

neurological function, suboccipital decompressive craniectomy should be performed

[321-323].Although simple ventricular drainage has the risk of tentorial notch hernia, if

cerebellar infarction causes significant brain oedema or mass effect, the risk can be

minimized by conservative cerebrospinal fluid drainage or suboccipital

decompressive craniectomy. Research data support cerebellar decompressive

craniectomy in patients with mass effect in acute ischemic minor stroke [321-323]. For

patients with cerebellar infarction, when it is impossible to treat obstructive

hydrocephalus with medication or ventricular drainage and neurological deterioration

due to cerebral oedema occurs, cerebellar decompressive craniectomy can be used for

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 89

intervention treatment [321-322].

Please refer to Figure 5 for a summary of the treatment process of cerebral

oedema/intracranial hypertension.

Recommendations

[1] Patients with major infarctions are at high risk developing brain edema and

intracranial hypertension. Transfer of patients to intensive care unit should be

considered. Measures to lessen the risk of edema and close monitoring of patients

for signs of neurological worsening during the first days after stroke are

recommended (Class I, Level of Evidence C).

[2] In patients ≤60 years of age with unilateral MCA infarctions who deteriorate

neurologically within 48 hours despite medical therapy, decompressive

craniectomy with dural expansion is reasonable (Class IIa, Level of Evidence A).

[3] In patients >60 years of age with unilateral MCA infarctions who deteriorate

neurologically within 48 hours despite medical therapy, decompressive

craniectomy with dural expansion may be considered (Class IIB, Level of

Evidence B).

[4] Although the optimal trigger for decompressive craniectomy is unknown, it is

reasonable to use a decrease in level of consciousness attributed to brain swelling

as selection criteria (Class IIa, Level of Evidence A).

[5] Ventriculostomy is recommended in the treatment of obstructive

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 90

hydrocephalus after a cerebellar infarct. Concomitant or subsequent

decompressive craniectomy may or may not be necessary on the basis of factors

such as infarct size, neurological condition, degree of brainstem compression,

and effectiveness of medical management (Class I, Level of Evidence C).

[6] Decompressive suboccipital craniectomy with dural expansion should be

performed in patients with cerebellar infarction causing neurological

deterioration from brainstem compression despite maximal medical therapy.

When deemed safe and indicated, obstructive hydrocephalus should be treated

concurrently with ventriculostomy (Class I, Level of Evidence B).

[7] Use of salvage osmotic therapy for patients with clinical deterioration from

occupying signs associated with major supratentorial infarction or cerebellar

infarction is reasonable (Class IIa, Level of Evidence C).

[8] Use of brief moderate hyperventilation (PCO2 target 30–34 mm Hg) is a

reasonable treatment for patients with acute severe neurological decline from

brain swelling (Class IIa, Level of Evidence C).

[9] Hypothermia or barbiturates in the setting of ischemic cerebral or cerebellar

swelling is not recommended (Class III, Level of Evidence B).

[10] Because of a lack of evidence of efficacy and the potential to increase the

risk of infectious complications, corticosteroids (in conventional or large doses)

should not be administered for the treatment of cerebral edema and increased

intracranial pressure (Class III, Level of Evidence A).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 91

(II) Haemorrhagic transformation

Ischemic stroke is often accompanied by petechial haemorrhage, patients have not

received vascular recanalization treatment and has no deterioration of neurological

function related to haemorrhage [324-325]. However, symptomatic haemorrhage occurs

in 5%-6% of patients treated with rt-PA intravenous thrombolysis, intravascular

recanalization and anticoagulant drugs [326-329].Haemorrhagic transformation can also

occur in patients who have not undergone vascular recanalization therapy, especially

in patients with massive infarction, older age, cardiogenic embolism and other factors.

Studies show that the prognosis of asymptomatic haemorrhagic transformation

patients is not different from that of patients without haemorrhagic transformation,

and there is still no research evidence for its treatment. Therefore, there is currently no

recommended special treatment for asymptomatic haemorrhagic transformation

patients. Symptoms and signs of symptomatic haemorrhagic transformation are

similar to spontaneous cerebral haemorrhage, such as deterioration of nervous system

function, mental state decline, headache, increase of blood pressure and pulse, and

vomiting [330]. Therefore, early clinical judgment, early warning and adjustment of

treatment plan are of great significance to the prognosis of these patients. At present,

there are many predictive scoring models for haemorrhagic transformation risks,

which can help guide the expectations of patients and their families, and may indicate

the required medical monitoring intensity of individual patients after intravenous

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 92

thrombolytic therapy. Treatment of haemorrhagic transformation requires complex

comprehensive management, including blood pressure management, reversal of

coagulation dysfunction and management of complications such as intracranial

hypertension [331]. At present, there is also a lack of high-quality research evidence on

symptomatic haemorrhagic transformation therapy and when to reuse antithrombotic

drugs (anticoagulant and antiplatelet), so there is no unified standardized therapeutic

regimen. A large number of observational studies show that the initiation or

continuation of antithrombotic therapy for patients with haemorrhagic transformation

after AIS will not lead to deterioration of the disease condition, provided that

assessment of the clinical indications, benefits and risks is ensured [332-333].

See Supplemental Table 9 for specific types of haemorrhagic transformation in

ischemic stroke.

Please refer to Figure 6 for details of the treatment process of haemorrhagic

transformation of acute ischemic stroke.

Recommendations

[1] Symptomatic haemorrhagic transformation: stop using anti-thrombotic

(antiplatelet, anti-coagulation) (Class I, Level of Evidence C); For haemorrhage

management associated with anticoagulation and thrombolysis, refer to cerebral

haemorrhage treatment guidelines.

[2] For AIS patients with haemorrhagic transformation, starting or continuing

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 93

antiplatelet or anticoagulant therapy should only be decided according to the

specific clinical conditions and potential indications (Class IIb, Level of Evidence

B).

(III) Epilepsy after stroke

The incidence rate of epilepsy after ischemic stroke is quite different in various

literature reports, but the incidence rate in most cases is less than 10% [334-335]. Some

reports show that the incidence of epilepsy in patients with haemorrhagic

transformation of ischemic stroke is significantly increased [336]. The incidence of

recurrent and delayed epilepsy in previous reports is also quite different [337-338]. The

applicable data on the effectiveness of anti-epileptic drugs for stroke patients are

limited, and the current recommendation is based on the established management over

neurological diseases complicated with epilepsy. At present, there is no research

evidence showing that preventive antiepileptic therapy can be benificial after

ischemic stroke, and there is only a small amount of relevant information about the

indications of long-term use of antiepileptic drugs after epileptic seizure.

See Figure 7 for details of the treatment process of the first epileptic seizure within 24

hours after stroke onset.

Recommendations

[1] Recurrent seizures after stroke should be treated in a manner similar to when

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 94

they occur with other acute neurological conditions, and anti-seizure drugs

should be selected based upon specific patient characteristics (Class I, Level of

Evidence C).

[2] Prophylactic use of anti-seizure drugs is not recommended (Class III, Level of

Evidence B).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 95

Section 7 Early evaluation and diagnosis of aetiology and pathogenesis of

ischemic cerebrovascular disease

I. Recommended examination and evaluation to be performed

(I) Imaging examination of brain and blood vessels

1. Brain parenchyma imaging

(1) Noncontrast CT

1) According to a number of large sample clinical observational studies, plain CT is

an optimal cost-effective imaging method, which can detect intracranial haemorrhage

and avoid antithrombotic treatment in these patients [339-340]. MRI-DWI is more

sensitive than noncontrast CT for detecting AIS, especially for posterior circulation

ischemic stroke[341-343]. In many patients, the diagnosis of ischemic stroke can be

made on the basis of the clinical symptoms and noncontrast CT excluding intracranial

haemorrhage [344]. MRI-DWI can provide certain information for the follow-up

intravascular treatment in patients with suspected diagnosis of ischemic stroke or

unclear location of responsible vessel areas.

2) A number of large clinical trials, including NINDS, ECASS II, IST-3 and a meta-

analysis in 2016, showed that there was no statistically significant modification of the

effect of rt-PA by the following findings on baseline CT including hypodensity in

brain parenchyma, hyperdense middle cerebral artery sign, semi-quantitative score

ASPECTS or leukoaraiosis [345-351].

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 96

Recommendations

[1] All patients admitted to acute care hospital for ischemic stroke should be

evaluated by cranial imaging. For the majority of patients, the head CT scan is

the first selection (Class I, Level of Evidence B).

[2] Early ischemic signs or leukoaraiosis showed on CT cannot be an absolute

exclusion standard for IV thrombolysis (Class III, Level of Evidence B).

(2) MRI

Two meta-analyses in 2016 have shown that cerebral microbleeds (CMB) are risk

factors for symptomatic intracranial haemorrhage(sICH) after thrombolysis

(OR=2.18, 95% CI: 1.12-4.22, P=0.021; OR=2.36,95% CI: 1.21~4.61,P=0.01).

However, sICH in patients with baseline CMBs is not more common (6.1%, 6.5%)

than in the NINDS rtPA trial (6.4%) [77, 352]. At the same time, both meta-analyses

include observational studies.

To date, no relevant RCT have been conducted to confirm the effect of baseline CMB

on the treatment efficacy and clinical outcome of thrombolysis. Therefore, according

to the existing evidence, CMB on baseline MRI can not affect the clinical decision

about intravenous thrombolysis.

Recommendations

Routine use of MRI to identify intracranial microhemorrhage, which can affect

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 97

decisions to IV thrombolysis, is not recommended (Class III, Level of Evidence

B).

2. Intracranial and extracranial angiography

A systematic review of 36 studies in 2018 concluded that currently there is no

prediction instruments with high sensitivity and specificity (including NIHSS,

CPSSS, LAMS, RACE) to diagnose large vessel occlusion. The study believed that

the NIHSS score is still the best tool to evaluate the disease condition in emergency,

but there is no strong evidence for pre-hospital identification and transfer of patients

with large artery occlusion [353].

Many observational studies which inculde more than 100 AIS patient suggest that the

risk of contrast-induced nephropathy secondary to CTA imaging is relatively low,

particularly in patients without a history of renal impairment. Therefore, there is no

need to delay starting treatment in order to wait for creatinine results [354-356].

Knowledge of extracranial vessel anatomy and presence of dissections, stenoses or

occlusions may assist in evaluating the risk of endovascular treatment and making

treatment decision.

Recommendations

[1] Patients with suspicious endovascular treatment should complete the non-

invasive vascular evaluation as soon as possible, but should pay attention to

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 98

avoid delaying thrombolysis (Class I, Level of Evidence B).

[2] Patients with suspected arterial occlusion and without previous renal

impairment can conduct the head and neck CTA inspection directly, in order to

avoid delay in treatment time in order to wait for creatinine to be resulted (Class

I, Level of Evidence B).

[3] For patients who may need intravascular therapy, completing the evaluation

of extracranial vessels including extracranial internal carotid artery and

vertebral artery is helpful for guiding the treatment options (Class IIa, Level of

Evidence C).

(2) Cardiac examination (structure, rhythm, function and ECG activity)

1. Cardiac structure evaluation Besides atrial fibrillation (AF), cardiac structure and

function are associated with higher risk of stroke, including acute myocardial

infarction, ischemic and non-ischemic cardiomyopathy, valvular heart disease

(including prosthetic valves and infective endocarditis), patent foramen ovale and

atrial septal aneurysm (ASA), cardiac tumors, and aortic atherosclerosis.

A meta-analysis indicates that the risk of ischemic stroke after acute myocardial

infarction is 11.1/1000 (95% CI: 10.7-11.5) during hospitalization, 12.2/1000 (95%

CI: 10.4-14.0) within 30 days of onset, and 21.4/1000 (95% CI: 14.1-28.7) within 1

year of onset [357].The factors related to embolism are anterior wall infarction and left

ventricular thrombosis. Studies found that the incidence of left ventricular thrombosis

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 99

was 6%-15% in patients with anterior wall infarction, and about 27% in patients with

anterior wall infarction complicated with left ventricular ejection fraction < 40% [358-

360]. Systemic embolism will occur in about 11% of patients with left ventricular

thrombosis [361].

Stroke occurs at an annual rate of 1% in cardiomyopathy patients with sinus rhythm

[362-364]. Causes of cardiomyopathy include infection, heredity and valvular heart

disease, coronary artery disease, hypertension and tachycardia, which eventually lead

to myocardial dysfunction and even congestive heart failure [365].Left ventricular

dysfunction leads to mural thrombus and then causes embolism events.

The annual incidence of systemic embolism events in patients with rheumatic valvular

disease is 1.5%-4.7% [366]. Thrombosis and subsequent embolism events are more

likely due to enlarged left atrium. The reported conclusions on the association

between mitral valve prolapse and cerebral embolism are inconsistent [367-369]. A

population-based study in the United States found a higher risk of stroke and TIA

among patients with mitral valve prolapse in sinus rhythm (RR=2.2, 95% CI: 1.5~3.2)

[370], and independent risk factors were older age, mitral thickening and atrial

fibrillation. According to a data analysis from the Framingham Study, mitral annulus

calcification was associated with a relative risk of stroke (RR=2.1, 95% CI: 1.2~3.6)

[371],and independent risk factor was the severity of mitral annular calcification. An

Amerindian cohort study also suggested that mitral annulus calcification increased

stroke risk (RR=3.1, 95% CI: 1.8~5.2) [372]. In contrast, a multi-ethnic study found

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 100

that mitral annulus calcification was associated with myocardial infarction and

vascular death rather than ischemic stroke [373]. At present, there is no evidence that

anticoagulant therapy can reduce stroke risk in patients with mitral annulus

calcification. Embolism events caused by calcification of aortic stenosis are

uncommon unless interfered by valvuloplasty, transcatheter or surgical valve

replacement [374].

The heart valve prosthesis can be a source of embolus. Mechanical valves have higher

risk to embolize than biological valves. The incidence of embolic events among

biological valve patients in sinus rhythm is about 0.7% every year [375]. Biovalve-

related embolic events usually occur within 3 months after operation and are more

common in mitral valve than in aortic valve [376].

Embolism occurs in 20%-40% of patients with endocarditis, most of which causes

central nervous system damage [377-378]. Embolism events are greatly reduced after

anti-infection treatment [379-381]. The related risk factors were excrescence size, mitral

valve involvement and staphylococcus aureus infection[379-380, 382].

Primary benign tumours of the heart (such as myxoma and papillary fibroma) and

primary malignant tumours (such as sarcoma) can form emboli and cause ischemic

stroke [383-384]. Tumours with fragile surfaces in the cardiac chamber are more prone to

embolism. Myxoma is the most common cardiac tumour, and is often found in the left

atrium [385]. 30%-40% of myxoma will be complicated with embolism [386]. Stroke and

TIA are the first symptoms in half of cardiac papillary fibroma patients [387-388].

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 101

Aortic atherosclerotic plaques ≥ 4mm, especially large and complex plaques, are

associated with the increased risk of cryptogenic stroke [388-392]. A French study found

that aortic atherosclerosis plaques > 4 mm can independently predict stroke

recurrence (RR=3.8, 95%CI: 1.8~7.8) [393]. In the PICSS study, it was found that large

plaques detected by transoesophageal echocardiography were associated with a

significantly increased risk of recurrent stroke or death during a 2-year follow-up

(RR=6.42, 95%CI: 1.62-25.46), which also existed in patients with large complex

plaques (RR=9.50, 95%CI: 1.92~47.10) [390]. Atherosclerotic embolism caused by

atherosclerotic plaques is the cause of stroke associated with cardiac surgery [390-392,

394].

Various types of cardiac surgery are also important risk factors for embolism. Cardiac

surgery such as valve replacement, coronary artery bypass grafting, radiofrequency

ablation and labyrinthine surgery had a high incidence of perioperative embolism.

Embolic sources can be either the operation itself or the implants. And embolism is

mostly related to secondary atrial fibrillation [395-401]. Anticoagulant therapy during

perioperative period can improve prognosis[441].

Among the common cardiac structure assessment methods, ordinary chest X-ray

examination can observe the atrioventricular enlargement and pulmonary oedema

through mediastinum. Transthoracic ultrasonography can better detect left ventricular

thrombosis than transoesophageal ultrasonography, while transoesophageal

ultrasonography can better reflect left atrial appendage thrombosis and patent foramen

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 102

ovale[403]. Transoesophageal ultrasonography can also identify left atrial appendage

morphology, which is associated with embolus formation [404]. Cardiac magnetic

resonance can further show potential embolic sources that cannot be shown by cardiac

cardiac ultrasonography. In a prospective study, about 21% of patients originally

considered as cryptogenic stroke were redefined as cardiogenic embolism after

routine examination. [405]. Cardiac magnetic resonance is very sensitive to left

ventricular thrombosis, and can also detect embolic sources such as ventricular

noncompaction insufficiency, atrial fibrosis, and complex atherosclerotic plaque [406-

409].

Recommendation:

[1] All stroke patients should complete routine chest X-ray and transthoracic

echocardiography in order to detect possible cardiac structural diseases (Class I,

Level of Evidence C).

[2] For stroke patients with suspected cardiogenic embolism, performing

transesophageal ultrasonography to determine the presence of left auricular

thrombosis, PFO or interatrial septal aneurysm is reasonable (Class IIa, Level of

Evidence B).

[3] Transthoracic echocardiography cannot be replaced by transesophageal

echocardiography (Class III, Level of Evidence C).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 103

[4] Cardiac MRI is effective in identifying the etiology of cryptogenic stroke. It

can be carried out in hospitals which can do cardiac MRI (Class IIb, Level of

Evidence B).

[5] Specific heart lesions found during cardiac screening in stroke patients

should be actively guided in specialized individual treatment (Class I, Level of

Evidence B).

2. Cardiac rhythm assessment

It is reported that the incidence of atrial fibrillation in Asian population is lower than

that of white people (~1% vs. ~2%), but the overall disease burden is still high due to

the aging population [410-411]. Even if there are no other cardiac diseases, atrial

fibrillation induces thrombus in left atrial appendage and increases the stroke risk by

4-5 times[412]. Embolism caused by atrial fibrillation accounts for more than half of all

cerebral embolism events. Asymptomatic atrial fibrillation and arrhythmia are very

common [413-415].A cohort study shows that about half (47%) of atrial fibrillation

patients were diagnosed after ischemic stroke [416].Patients with cardiogenic cerebral

embolism caused by atrial fibrillation have larger infarction area and higher

haemorrhagic transformation rate and mortality rate, causing greater nursing burden

[417-425].

Different from atrial fibrillation, the relationship of paroxysmal supraventricular

tachycardia, atrial flutter, sick sinus syndrome with stroke is unknown. Some studies

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 104

have reported that the annual incidence of stroke in patients with paroxysmal atrial

fibrillation, supraventricular tachycardia was similar to patients with persistent atrial

fibrillation, ranging from 1.5% to 3.3% [426-431]. Another retrospective cohort study

found that paroxysmal supraventricular tachycardia was associated with ischemic

stroke [432]. Other arrhythmias associated with increased risk of stroke have also been

reported, such as Bayes syndrome, and intra-atrial block. To date, there is no evidence

to prove that intervention on arrhythmia can reduce the occurrence of stroke[433-436].

Stroke risk assessment tools for patients with atrial fibrillation can effectively predict

the occurrence of clinical adverse events based on specific risk factors and can guide

treatment decisions. [437]. CHADS2 has been proved to be a simple and effective tool

for stroke risk assessment in patients with atrial fibrillation. Fully validated

independent risk factors for stroke in atrial fibrillation are used for assessment [81,438].

The full score is 6. Specifically, congestive heart failure, hypertension, age > 75,

diabetes each score 1 point, prior stroke or TIA scores 2 points. This score has been

confirmed in many cohort studies of atrial fibrillation. The stroke risk of patients with

atrial fibrillation is divided into the high-risk group (≥ 2 points, 1.9%-7.6%/year),

moderate-risk group (1 point, 1.2%-2.2%/year) and low-risk group (0 point, 0.5%-

1.7%/year) according to CHADS2 [438-441]. CHA2DS2-VASc sets vascular diseases

(peripheral vascular disease, myocardial infarction, aortic plaque), aged 65-75 years

and women each scores 1 point, which is more sensitive to embolic events than

CHADS2 score [442-443].The CHA2DS2-VASc score is a good assessment tool for

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 105

stroke and TIA risk in patients with atrial fibrillation, and is applicable even in

patients with end-stage renal disease and heart failure without atrial fibrillation [444-

445]. The absolute risk of stroke was included in the risk assessment, which is of

guiding significance for the formulation of complex drug interventions.

Studies showed that the detection rate of undiagnosed atrial fibrillation can be

increased in patients aged >65 years who are admitted to primary health care

institutions through professional pulse assessment. [446-447]. Routine systematic pulse

examination in the outpatient department and 12-lead ECG examination for patients

with irregular pulse rhythm can increase the diagnostic rate of atrial fibrillation by

60% [446]. The application of long-term ECG monitoring in cryptogenic stroke is

mainly used to detect paroxysmal atrial fibrillation. Continuous ECG monitoring,

repeated ECG examination and 24h Holter monitoring are the first-line diagnostic

procedures within 48 to 72h after the onset of stroke. If embolism is suspected and no

other reasons are found, there are various methods to prolong ECG monitoring, such

as implanting a loop recorder in vitro or in vivo, or reading pacemaker or defibrillator

data through external telemetry, etc. [448-449]. Studies showed that extending the

monitoring time of heart rhythm to 10 days, 30 days or even 6 months will greatly

increase the detection rate of paroxysmal atrial fibrillation. But at the same time, the

risk of over-diagnosis will increase, because only paroxysmal atrial fibrillation

exceeding 30 seconds is meaningful to embolism events [450-452]. The TRENDS study

reported that in the long-term monitoring, patients with burden of atrial fibrillation or

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 106

atrial tachycardia > 5.5h within 30 days had a doubled risk of cerebral and systemic

embolism events [453]. The trial of portable examination equipment for people at a

high risk of atrial fibrillation is underway and the results are expected [454]. Atrial

fibrillation may runs in family, and genetic screening for patients with high risk for

atrial fibrillation may be possible in the future [455].

Recommendations

[1] Asymptomatic atrial fibrillation and arrhythmia are very common, screening

for atrial fibrillation should be routinely performed in the clinic, the routine

checking of pulse should be performed on a patient > 65 years old, and a 12-lead

ECG should be conducted on patients with abnormalities of pulses (Class I,

Level of Evidence A).

[2] The CHADS2 or CHA2DS2 - VASc score is recommended for patients with

persistent atrial fibrillation when assessing for the risk of stroke, and used to

guide intervention (Class I, Level of Evidence A).

[3] In patients with latent stroke who may have embolism, 24h or long term and

remote cardiac monitoring aiming to find any paroxysmal atrial fibrillation is

reasonable (Class IIa, Level of Evidence B).

[4] For patients with non-persistent atrial fibrillation or paroxysmal atrial

fibrillation/atrial tachycardia (> 5.5h) within 30 days or paroxysmal atrial

fibrillation for more than 30 seconds, stroke prevention treatment as in patient

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 107

with persistent atrial fibrillation may be reasonable (Class IIb, Level of Evidence

B)

[5] Whether arrhythmias other than atrial fibrillation or paroxysmal

supraventricular tachycardia are associated with embolic events is unclear, and

any intervention of those arrhythmias to reduce the incidence of embolism is still

inadequate, symptomatic treatment can be taken into consideration (Class III,

Level of Evidence C).

3. Cardiac function assessment

Cardiac dysfunction is usually secondary to organic heart disease or various

arrhythmias that affect myocardial coordination. Even in sinus rhythm patients

without left atrial enlargement [456-458], decreased blood flow velocity in left atrium

and left atrial appendage also increases the risk of embolic stroke. In patients with

atrial fibrillation, the stroke risk increases as the left atrium enlarges and the left atrial

emptying fraction decreases. Even in patients with normal left atrium size and sinus

rhythm, left atrial dysfunction also exists and the risk of stroke is increased [459]. Left

atrial spontaneous echo contrast (LASEC) is associated with thrombosis, and

rheumatic heart disease patients with LASEC have a higher CHA2DS2 - VASc

average than patients without LASEC [460]. Patients in sinus rhythm have moderate or

severe decrease of left ventricular systolic function (EF ≤ 35%) and a flow rate of left

atrial appendage ≤ 55cm/s, which is easy to induce LASEC, and they are high risk

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 108

populations for stroke [461].Von Willebrand factor (vWF) plays a role in left atrial flow

velocity reduction and thrombosis, and is relatively high in plasma of patients with

atrial fibrillation [462-463].For ischemic stroke patients complicated with paroxysmal

atrial tachycardia, the enlarged left atrial volume index is an independent risk factor

for atrial fibrillation, which can predict the recurrence of stroke. It is suggested that

24h Holter monitoring and appropriate anticoagulant therapy be performed for the

above patients [464].

Recommendations

[1] It is recommended to include cardiac function in the cardiac assessment of

stroke patients: the left atrium, left atrium, left ventricle function, specific

projects including volume index, emptying index and blood flow velocity (Class I,

Level of Evidence B).

[2] The left atrium, left auricle and left ventricular blood flow disturbance, and

left atrial spontaneous ultrasound contrast phenomenon are independent risk

factors for embolus formation and triggering embolism. It is necessary to find

the cause and take intervention measures actively (Class IIa, Level of Evidence

B).

(III) Examination and assessment of cryptogenic stroke

About 80% of patients with cryptogenic stroke are neither related to lacunar (caused

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 109

by arteriolar diseases) nor severe atherosclerotic stenosis, and have no major source of

cardiac embolism (such as atrial fibrillation). A recent monitoring and imaging study

showed that most of these patients had potential embolic sources, suggesting that a

large proportion of cryptogenic stroke in the past can be described as embolic stroke

of undetermined source (ESUS) [465]. ESUS patients are embolic stroke and their

major risks of cardiogenic embolism, occlusive atherosclerotic stroke and lacunar

stroke are excluded by sufficient diagnostic assessment. Potential causes of ESUS

include low-risk potential cardiac embolism [myxomatous valve prolapse, mitral

valve calcification, aortic stenosis, calcified aortic valve, cardiac arrest, sick sinus

syndrome, atrial fast rhythm, left atrial appendage stagnation with reduced blood flow

velocity, spontaneous hypoecho, atrial septal aneurysm, Chiari network, moderate

systolic or diastolic dysfunction of left ventricle (diffuse/focal), ventricular non-

contraction and endocardial myocardial fibrosis], latent paroxysmal atrial fibrillation,

arterial embolism (atherosclerotic plaque in the aortic arch and cerebral artery non-

stenotic ulcer plaque), abnormal embolism (patent foramen ovale, atrial septal defect

and pulmonary arteriovenous fistula), tumor-related and unknown causes. See Figure

8 for the diagnostic flow of ESUS.

II. Risk factor assessment and risk stratification

(I) Blood pressure assessment

About 80% of patients have elevated blood pressure after AIS. The blood pressure

level of patients and the timing of initiating antihypertensive therapy for patients are

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 110

closely related to prognosis. Therefore, blood pressure assessment after onset is of

great significance.

In terms of baseline blood pressure after ischemic stroke, some studies have shown

that higher baseline blood pressure is related to good prognosis, while low blood

pressure variability in the early phase (within 72 hours) is independently related to

good prognosis [466-472]. In addition, the study showed that the relationship between

blood pressure and prognosis was due to U-shaped effect. With a limit of

180/100mmHg, patients with the highest or lowest values of SBP and DBP had a

higher frequency of adverse prognosis [473]. For patients with culprit artery stenosis

less than 50%, elevated pulse pressure at the early stage is independently associated

with unfavorable late outcome[474]. And follow-up of a large sample study showed a J-

shaped effect between pulse pressure and recurrence of cardiovascular events [475]. For

patients with severe arterial stenosis, antihypertensive therapy is associated with

stroke progression and poor prognosis [476].

Recommendations

[1] Hypertension after AIS should be moderated strictly and lowered moderately,

controlling the blood pressure in 140~160/80~99 mmHg within 24~48h is

reasonable (Class I, Level of Evidence A).

[2] Blood pressure variation and pulse pressure should be monitored closely after

AIS. Blood pressure correlates the prognosis (Class IIa, Level of Evidence B).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 111

(II) Assessment of blood lipid

Assessment of blood lipid after AIS is helpful to judge the pathogenesis and guide the

treatment in the secondary prevention of ischemic stroke. At present, various reports

suggest that blood lipid should be controlled at the low level of normal range,

especially the relatively high level of low-density lipoprotein which requires intensive

lipid-lowering therapy (the target value for lowering is set as LDL-C concentration <

1.8mmol/L or > 50% lower than baseline). A large sample size randomized controlled

study showed that intensive lipid-lowering therapy can reduce the recurrence of stroke

after ischemic stroke, but may increase the risk of haemorrhage [477],and many small

sample studies also confirmed this result. [478-481]. Another large sample size cohort

study also found that high-dose atorvastatin increased the risk of haemorrhage, but

was not correlated with baseline or recent LDL-C [482]. However some small sample

studies found that [483-485] the lower baseline blood lipid at admission indicates that

cerebral infarction was more serious [486-487], and dyslipidaemia (high or low) was

associated with poor prognosis [488-491]. Meanwhile, a large sample size multi-center

cross-sectional study showed that the up-to-standard rate of blood lipid after stroke ( <

1.8mmol/L) was still relatively low, which was about 27.4% in China, and drug

therapy and health education still need to be vigorously promoted [492-495].

Recommendations

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 112

[1] Dyslipidemia (too high or too low) is closely related to poor prognosis. Serum

lipid level should be actively assessed after AIS, in order to guide lipid-lowering

treatment and secondary prevention (Class IIa, Level of Evidence B).

[2] Relatively low blood lipids may indicate that the condition of cerebral

infarction is more serious, and attention should be paid to the changes of

patients' condition (Class IIb, Level of Evidence C).

[3] At present, the rate of reaching the standard of blood lipid control after AIS

in China is still low, blood lipid control should be strengthened, at the same time

pay attention to regular monitoring to avoid bleeding transformation (Class I,

Level of Evidence B)

(III) Assessment of abnormal glucose metabolism

Many studies and meta-analysis have shown that hyperglycaemia (≥ 7.7mmol/L) after

stroke, history of diabetes and high blood glucose fluctuation were closely related to

low recanalization rate after thrombolysis and worse clinical outcome [534-554]. The

blood glucose level may indicate the severity of the disease [496-497]. However, β-cell

dysfunction was associated with an increased risk of poor prognosis in nondiabetic

patients with ischemic stroke[498]. Some small-sample RCT showed that it was

feasible and tolerable to inject insulin within 48 hours to control blood glucose strictly

(5-8 mmol/L), which can improve stroke prognosis [499-500]. A small-sample RCT

found that intravenous infusion of glucose-insulin-potassium solution can reduce

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 113

blood glucose and improve the brain lactic acid level, but cannot improve stroke

progression [501]. Some studies have also shown that insulin pumping for

hypoglycaemic therapy after stroke and health education for diabetic patients can

benefits from controlling blood glucose. The incidence of hypoglycaemia after stroke

is relatively low. But if it occurs, it will directly aggravate cerebral ischemia injury

and cerebral ooedema, leading to poor prognosis. Therefore, blood glucose should be

closely monitored to prevent hypoglycaemia.

Recommendations

[1] High blood sugar level and blood sugar fluctuation after AIS is closely related

to the prognosis. Clinical monitoring and strict glycemic control are

recommended (Class I, Level of Evidence A).

[2] Blood glucose should be strictly monitored after AIS and insulin should be

recommended for stable hyperglycemic control, it is reasonable to control the

blood glucose concentration to 5 ~ 8 mmol/L (Class IIa, Level of Evidence B).

(IV) Risk stratification assessment of TIA

1. California risk score In 2000, a cohort study of 1707 TIA patients showed that

180 patients had recurrent stroke within 90 days. From the data, independent risk

factors were screened through multivariable logistic regression analysis, and

California risk score was established: A. Age > 60 years; B. Diabetes; C. Duration of

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 114

symptoms > 10min; D. Symptom of physical weakness; E. Speech impairment. It is

used to predict the short-term stroke risk of TIA patients and the highest score is 5.

2. SPI-I and SPI-Ⅱ risk scores In 1991, Kernan et al established the SPI-I score and

scored and followed up 142 TIA and minor stroke patients to predict the risk of stroke

and death within 2 years after symptoms onset. The score included: A. Age > 65 (3

points); B. Diabetes (3 points); C. Blood pressure > 180/100 mmHg (2 points), D.

Coronary atherosclerotic heart disease (1 point); E. The first event was stroke or TIA

(2 points). The highest score was 11. In low risk group (0-2 points), intermediate risk

group (3-6 points) and high-risk group (7-11 points), corresponding outcome rates

were 3%, 27%, and 48% in the original cohort and 10%, 21%, 59% in the test cohort

respectively.

Then, on the basis of the SPI-I score, past stroke history (3 points) and congestive

heart failure (3 points) were added to establish the SPI-II score [502], with a maximum

of 15 points, to evaluate the incidence of stroke or death within 2 years.

3. Essen stroke risk score Essen Stroke Risk Score (ESRS) is developed based on

the CAPRIE test database and is one of the few prediction tools for judging stroke

recurrence risk based on the ischemic stroke population. Its content includes: A. 65-

75 years old; B. > 75 years old; C. Hypertension; D. Diabetes; E. Previous myocardial

infarction; F. Other cardiovascular diseases; G. Peripheral arterial diseases; H.

Smoking; I. Previous TIA or ischemic stroke; Specifically, except for B. > 75 years

old whose score is 2 points, the others score 1 point, and the highest score is 9.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 115

Patients are divided into the low risk group (0-2) and high-risk group (≥ 3). At

present, the score is also applied to predict the recurrence risk in patients with TIA

and minor stroke [503].

4. ABCD score system California, SPI-I, SPI-II and Essen scores all predict long-

term prognosis. However, recurrent stroke often occurs in a short period of time for

TIA patients. Therefore Rothwell et al. established the ABCD score to predict stroke

risk within 7 days after TIA, and on this basis, ABCD2, ABCD2-MRI, ABCD2-I,

ABCD3 and other scores were established.

(1) ABCD score [567]: Based on the Oxford Shire Community Stroke Project (OCSP),

ABCD score was established to predict the risk of stroke within 7 days after TIA,

with a total score of 6, including:

A. Age ≥ 60 (1 point).

B. Blood pressure: SBP > 140mmHg and/or DBP ≥ 90mmHg (1 point).

C. Clinical manifestations: unilateral limb weakness (2 points), speech disorder

without limb weakness (1 point), other symptoms (0 point).

D. Symptoms lasting for > 60 min (2 min), 10-59 min (1 point), and < 10 min (0

point).

The predictive value of the ABCD score was validated in OXVASC study. The results

showed that the 7-day risk of stroke was 0.4% in patients with an ABCD2 score of

more than 5, 12.1% with a score of 5, and 31.4% with a score of 6. It suggested that

the higher the ABCD score of TIA patients, the higher the risk of stroke within 7 days.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 116

Rothwell et al. suggested that patients with ABCD score ≤ 4 generally do not need

hospitalization observation, while patients with a score of 6 are in the acute phase of

disease and need to be hospitalized for observation and treatment [504].

(2) ABCD2 score: Based on the California risk score and ABCD score, ABCD2 score

was proposed to predict the risk of stroke within 2 days after TIA. Compared with the

ABCD score, the ABCD2 score increases the risk factor of diabetes (1 point), with a

total score of 7 points. At the same time, the stroke risks of 2,893 TIA patients in 4

groups of cohorts were predicted for 2 days, 7 days, 30 days and 90 days. The results

showed that the stroke risks of the high-risk group (6-7 points), intermediate risk

group (4-5 points) and low risk group (0-3 points) within 2 days after TIA were 8.1%,

4.1% and 1.0% respectively, indicating that the ABCD2 score can be used to predict

the early stroke of TIA patients. However, ABCD2 has limited effect on the

assessment of early recurrent stroke in patients with mild stroke. This result is based

on the Oxford Vascular study that evaluated the predictive value of ABCD2 and other

scoring systems for 7d and 90d recurrence risk of patients with mild stroke (NIHSS ≤

5). The study results showed that the predictive value of ABCD2 was of a medium

level [505].

(3) ABCD2-MRI and ABCD2-I score: With the rapid development of imaging

technology, CT and MRI have become an important method for clinical diagnosis and

prognosis evaluation of cerebrovascular diseases. Combining imaging markers with

the ABCD scoring system can improve the prediction level.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 117

In 2008, researchers established the ABCD2-MRI score based on the ABCD2 score

and imaging markers. The score added the intracranial artery stenosis and DWI high

signal, each with a score of 1. 180 TIA or mild stroke patients were examined by head

MRI (images within 24 hours after onset) and scored. The results showed that 11.1%

of the patients had recurrent stroke events within 90 days after onset. Specifically, the

recurrent stroke rates for 90 days in the high-risk group (7-9 points), intermediate risk

group (5-6 points) and low risk group (0-4 points) were 32.1%, 5.4% and 0.0%

respectively. Meanwhile, the rate of dysfunction within 90 days was 22.9%, 7.5% and

7.7% respectively. However, the ABCD2 score could not predict dysfunction of the

NICE patients. Therefore, based on clinical and MRI information within an effective

time window, it is possible to more accurately predict the risk of recurrent stroke after

TIA or minor stroke. In 2010, the ABCD2-I score added DWI high signal based on

the ABCD2 score and assigned 3 points to it, and 4,574 TIA patients were predicted

for stroke risk. The results showed that the ABCD2-I score improved the predictive

value for stroke risk within 7 days and 90 days after TIA compared with ABCD2 [506].

(4) ABCD3 and ABCD3-I score: The ABCD3 score was proposed according to the

ABCD2 score[507].The ABCD3 score adds two factors to the ABCD2 score: treatment

of TIA patients within 7 days before onset and occurrence of TIA at least once before,

with a total score of 0-9 points. Researchers found that the ABCD3 score and ABCD2

score have similar predictive value for stroke recurrence risk within 7 days and 90

days after TIA. However, the ABCD3 score can not to be wide use yet because the

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 118

validity test has not been conducted.

Meanwhile, on the basis of the ABCD3 score, ipsilateral carotid stenosis and DWI

abnormal high signal were added in ABCD3-I score. It was established to predict the

stroke risk of 886 TIA patients. The results showed that the ABCD3-I score improved

the prediction accuracy compared with the ABCD2 score.

(5) ABCDE+ score: Recently, some researchers added etiological classification and

imaging findings to the ABCD2 score and established the ABCDE+ score. This is the

first score that added etiological classification into the ABCD scoring system. In 248

TIA models, the AUC value of the ABCDE+ score was higher than that of the

ABCD2 score (P=0.04). However, this score has not been widely accepted yet.

Among the many risk models, the ABCD scoring system is the most widely used.

Specifically, the ABCD2 score can well predict the short-term stroke risk of TIA

patients, and is the most widely used. Its predictive value has been verified, and it has

been recommended by the expert consensus in China. The ABCD2-I, ABCD2-DWI

and ABCDE+ scores have rarely been verified in Chinese population. The ABCD3

and ABCD3-I scores are rarely used, which can more effectively assess the short-term

stroke risk of TIA patients.

(V) Stratified assessment of ischemic stroke

1. Atherosclerotic cardiovascular disease (ASCVD) The definition of clinically

confirmed ASCVD includes:

(1) Acute coronary syndrome

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 119

(2) History of myocardial infarction

(3) Stable or unstable angina pectoris

(4) Coronary artery or other vascular reconstruction

(5) Atherosclerotic stroke or TIA

(6) Atherosclerotic peripheral arterial disease

2. Essen stroke risk score [503](Supplemental Table 10) It is used to assess the risk

of stroke recurrence in patients with non-cardiogenic ischemic stroke, thus giving

individualized secondary prevention measures to different patients. The total score is

9 points, 0-2 for low risk, 3-6 for intermediate risk and 7-9 for high risk.

3. Risk stratification assessment of atrial fibrillation

(1) CHADS2 score and CHA2DS2-VASc score [508-510](Supplemental Table 11) : It

is mainly used to assess the risk of stroke in patients with atrial fibrillation and guide

antithrombotic therapy.

Classic CHADS2 score: Patients with the total score of more than 2 are high risk

group for stroke, and anticoagulant therapy is recommended. Patients with the total

score of 1~2 points are intermediate risk group, and anticoagulant therapy or

antiplatelet therapy can be selected, and anticoagulant therapy is recommended.

Patients with the total score of 0 are low risk group and antiplatelet therapy is

recommended.

Modified CHADS2 score: Lip et al. modified the stratification standard as 0 for low

risk, 1 for intermediate risk and 2-6 for high risk.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 120

The CHA2DS2-VASc score adds the risk factor of gender and refines the age

stratification: 0 point for the low risk group, 1 for the intermediate risk group and 2--6

for the high-risk group.

See Supplemental Table 12 for a summary of stratification assessment of stroke risks

for patients with atrial fibrillation.

(2) HAS-BLED scale [511]: It is mainly used to assess the bleeding risk of

anticoagulant therapy for patients with atrial fibrillation. The scale has a total score of

9 points, with a score ≥ 3 for high risk, 1-2 for intermediate risk, and 0 for low risk

(Supplemental Table 13).

When the CHADS2 score is ≥ 2, the patients have a high risk of stroke, and

anticoagulant therapy is recommended. If the HAS-BLED score is higher than

CHADS2 score, the bleeding risk exceeds the anticoagulant benefit. For patients with

CHADS2 score =1, the difference between the HAS-BLED score and CHADS2 score

is no more than 2 points, and the anticoagulant benefit is greater than the bleeding

risk.

4. SPI-Ⅱ scale SPI-Ⅱ is used to predict the risk of long-term stroke recurrence.

However, the scale is of little value in predicting the risk of stroke recurrence within

90 days after the minor stroke (NIHSS score < 3). The total score is 15 points, 0-3 for

low risk, 4-7 for intermediate risk, and 8-15 for high risk (Supplemental Table 14).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 121

III. Diagnosis of aaetiology and pathogenesis

The aaetiology of ischemic stroke is complex and diverse, and it is usually difficult

for a single classification standard to include all subtypes of stroke. Reasonable and

feasible ischemic stroke classification is very important to evaluate therapeutic effects

in clinical trials. In the past, the classification systems developed by western countries

were all based on white race, and their main mechanisms of stroke were cardiogenic

embolism or extracranial atherosclerosis. However, these classification systems may

not be applicable to Asian populations. The typing proportion of ischemic stroke is

related to race. Stroke data from western countries show that cardiogenic embolism is

the most important cause of ischemic stroke. In addition, the incidence of extracranial

atherosclerotic stenosis in American population is significantly higher than that of

intracranial atherosclerosis.

After the classical TOAST classification [80,512] system, various derivatives have been

generated successively, such as SSS-TOAST classification[88], CCS-TOAST

classification[72], South Korea -TOAST classification, etc. But the Chinese ischemic

stroke subtype CISS classification, which is defined and developed independently by

China [513] is currently the most suitable classification method for the aaetiology and

pathogenesis of Chinese population. The classification process of the standard system

is divided into two steps: The first step is similar to the classical TOAST

classification, and causes are also divided into five types including large artery

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 122

atherosclerosis (LAA), cardiogenic stroke (CS), penetrating artery disease (PAD),

other causes (OE) and undetermined aaetiology (UE). Specifically, LAA is also

divided into aortic arch atherosclerosis and intra- and extracranial large arteries

atherosclerosis by site. In the second step, the pathogenesis of intracranial and

extracranial atherosclerotic cerebral infarction is classified into 4 types including

parent artery (plaque or thrombus) occluding penetrating artery, artery to artery

embolism, hemodynamic/impaired emboli clearance and multiple mechanism.

(I) Large artery atherosclerosis

In CISS classification, large artery atherosclerosis includes aortic arch atherosclerosis

and intra- and extracranial large arteries atherosclerosis.

1. Aortic arch atherosclerosis

(1) Acute multiple infarction lesions, especially involving bilateral anterior and/or

anterior and posterior circulations.

(2) No evidence of atherosclerosis of relevant intracranial or extracranial large

arteries (vulnerable plaques or stenosis ≥50% or occlusion).

(3) No evidence of potential cause of cardiogenic stroke (CS).

(4) No evidence of other aetiologies that can cause multifocal acute ischemic infarcts

such as vasculitidies, haemostatic disturbances, and tumorous embolism.

(5) Evidence of significant aortic arch atherosclerosis: aortic plaques 24 mm and/or

aortic thrombi, detected by high resolution magnetic resonance (HR-MRI)/ MRA

and/or transoesophageal ultrasound (TEE).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 123

2. Intra- and extracranial large arteries atherosclerosis

(1) Any distribution of acute infarcts (except isolated infarct in the territory of one

penetrating artery), with evidence of atherosclerosis involving intracranial or

extracranial large arteries (vulnerable plaques or stenosis ≥50%) that supply the area

of infarction.

(2) Concerning isolated penetrating artery territory infarct, the following

circumstance should also be included in LAA: with evidence of atherosclerotic plaque

(detected by HR-MRI) or any degree of stenosis in the parent artery (detected by

TCD, MRA, CTA, or DSA).

(3) No evidence of potential cardiac-origin embolic cause.

(4) Other possible causes have also been excluded.

(II) Cardiogenic stroke

1. Diagnostic criteria

(1) Acute multiple infarcts, especially involving bilateral anterior and/or anterior and

posterior circulations (including cortical infarcts) that have occurred closely in time.

(2) No evidence of atherosclerosis on relevant intracranial or extracranial large

arteries (vulnerable plaques or stenosis ≥50% or occlusion).

(3) No evidence of other aetiologies that can cause multifocal acute ischemic infarcts

such as vasculitidies, haemostatic disturbances, and tumorous embolism.

(4) Evidence of cardiac disease that has a potential for embolism.

(5) If the possibility of aortic arch atherosclerosis has been excluded, CS is definite.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 124

Otherwise, the category should be possible CS.

2. The potential lesions included under this category are: mitral stenosis, prosthetic

heart valve, myocardial infarction within the past 4 weeks, mural thrombus in the left

cavities, left ventricular aneurysm, any documented history of permanent or transient

atrial fibrillation or flutter with or without spontaneous echo contrast or left atrial

thrombus, sick sinus syndrome, dilated cardiomyopathy, ejection fraction <35%,

endocarditis, intracardiac mass, PFO plus in situ thrombosis, PFO plus concomitant

PE, or DVT preceding the brain infarction.

(III) Perforating artery disease

Acute isolated infarct in the territory of one penetrating artery caused by

atherosclerosis at the proximal segment of the penetrating arteries or lipohyalinotic

degeneration of arterioles is called penetrating artery disease (PAD). Diagnostic

criteria: (1) Acute isolated infarct in clinically relevant territory of one penetrating

artery, regardless of the size of infarct. (2) No evidence of atherosclerotic plaque

(detected by HR-MRI) or any degree of stenosis in the parent artery (detected by

TCD, MRA, CTA, or DSA). (3) With evidence of vulnerable plaques or stenosis

≥50% in ipsilateral proximal intracranial or extracranial large arteries, isolated

penetrating artery infarct is classified in undetermined aaetiology (UE; multiple

aaetiology). (4) With evidence of cardiac disease that has a potential for embolism,

isolated penetrating infarct is classified in UE (multiple aaetiology). ⑤ Other

possible causes has been excluded.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 125

(IV) Other aetiologies

Evidence of other specific diseases (e.g., vascular related disease, infective disorder,

inherited disease, haematological system disorder, vasculitis), that are relevant to the

index stroke and can be demonstrated by blood tests, cerebrospinal fluid (CSF) tests,

and vascular imaging. The possibility of LAA or CS has been excluded.

(V) Undetermined aaetiology

1. No evidence of any specific potential aaetiology that is clinically relevant to the

index stroke.

2. Multiple: Evidence of more than one potential cause, but difficult to determine

which was the relevant cause of the index stroke.

3. Unknown: No determined cause is responsible for the index stroke unless more

investigations would be performed.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 126

Section 8 Intervention on aaetiology and pathogenesis

I. Aortic atherosclerotic stroke

(I) Antithrombotic therapy

Antiplatelet drugs combined with risk factor control are the main drug treatment plans

for patients with aortic atherosclerotic stroke. Aspirin is currently the most commonly

used antiplatelet drug worldwide.

In recent years, the use of dual antiplatelet drugs in patients with aortic atherosclerotic

stroke is increasing, especially in patients with intracranial arterial stenosis. The

evidence mainly comes from the following research results: Stenting and Aggressive

Medical Management for Preventing Recurrent Stroke in Intracranial Stenosis

(SAMMPRIS) [514], Vitesse Intracranial Stent Study for Ischemic Stroke Therapy

(VISSIT) [515] and Warfarin-Aspirin for Symptomatic Intracranial Disease (WASID)

[516].

In 2011, the large-scale randomized study SAMMPRIS compared the effect of

intracranial arterial stenting and intensive drug therapy for symptomatic severe

intracranial arterial stenosis on reducing stroke recurrence and mortality. The patients

were divided into two groups. One group was treated with intensive drug therapy, and

the other group was treated through stenting in addition to intensive drug therapy.

Drug therapy included aspirin (325 mg/d) and clopidogrel (75 mg/d) for 90 days.

Meanwhile, major risk factors such as hypertension and hypercholesterolemia as well

as secondary risk factors such as diabetes, smoking, overweight and insufficient

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 127

exercise were managed through lifestyle adjustment. The results showed that the 30-

day stroke recurrence or death rate in the interventional treatment group was

significantly higher than that in the medical treatment group, and the incidence rate of

major endpoint events in the interventional treatment group was also significantly

higher after 1-5 years of follow-up. After correcting baseline characteristics,

Chaturvedi et al. found that stroke or vascular death within 30 days in patients with

intracranial arterial stenosis who took aspirin or warfarin orally in the WASID study

was 1.9 times that in patients who took dual antiplatelet therapy orally in the

SAMMPRIS study, thus providing theoretical support for intensified drug therapy.

Another evidence for dual antiplatelet therapy comes from "Clopidogrel Plus Aspirin

versus Aspirin Alone for Reducing Embolization in Patients with Acute Symptomatic

Cerebral or Carotid Artery Stenosis" (CLAIR). The results of this study suggested that

in the intracranial atherosclerosis subgroup, the number of microembolic signals

detected by transcranial Doppler (TCD) was reduced (31% vs. 54%) in patients

treated with clopidogrel combined with aspirin compared with patients treated with

aspirin alone. The subgroup analysis of "Clopidogrel in High-risk Patients With Acute

Non-disabling Cerebrovascular Events" (CHANCE) found that patients with

intracranial artery stenosis using dual antiplatelet drugs to prevent stroke recurrence

had a tendency for good prognosis at 90 days compared with patients using aspirin

alone [517]. For patients with stroke or TIA caused by severe intracranial arterial

stenosis (70%-99%), use of aspirin combined with clopidogrel (75mg/d) for 90 days

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 128

may be reasonable.

Patients with stable coronary heart disease are high-risk groups for stroke, myocardial

infarction and vascular death. For a long time, although aspirin has been used as the

standard antithrombotic therapy, there have still been a considerable number of

cardiovascular events. Recently, breakthroughs have been made in the research and

development of oral factor Xa inhibitors. Previous studies have shown that aspirin or

other antiplatelet drugs combined with low-dose can be beneficial for

patients with acute coronary syndrome. Deeper understanding of whether low-dose

rivaroxaban combined with aspirin will further reduce the residual risk of

cardiovascular events in patients with coronary heart disease is needed.

The COMPASS study [518] is the first randomized, double-blind, multicentre,

prospective and randomized Phase III clinical study to evaluate the efficacy and safety

of new oral anticoagulants in the high-risk patient population of coronary artery

disease (CAD) or peripheral artery disease (PAD). A total of 27,400 CAD or PAD

patients were enrolled in the study and randomly divided into 3 groups. The present

treatment group included 2 groups: Rivaroxaban 2.5mg orally bid, plus aspirin 100mg

bid; Rivaroxaban 5mg orally bid; Aspirin 100mg orally qd. The primary efficacy

endpoint of the study was first occurrence of cardiovascular death, myocardial

infarction and stroke, the primary safety endpoint was massive haemorrhage, and

secondary endpoints included compound myocardial infarction, stroke, cardiovascular

death, venous thromboembolism, cardiovascular hospitalization and all-cause death.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 129

The results of the COMPASS study showed that for patients with coronary heart

disease and PAD who did not need dual antiplatelet therapy, combined application of

the new anticoagulant rivaroxaban and aspirin was significantly better than aspirin

alone in preventing cardiovascular events and could further reduce the risk of residual

cardiovascular events in this high-risk group. The subgroup analysis results showed

that combined application of rivaroxaban and aspirin could reduce stroke by 42%

compared with aspirin alone. Therefore, new and more specific measures of oral

anticoagulation combined with aspirin to prevent stroke are expected to be obtained

through subgroup analysis, and the study conclusion needs further tests to confirm.

Recommendations

[1] For patients with symptomatic intracranial artery stenosis, antiplatelet

therapy should be started as soon as possible and used long term. The alternative

antiplatelet drugs are aspirin, clopidogrel, and cilostazol. (Class I, Level of

Evidence A).

[2] Minor stroke patients with high-risk intracranial artery stenosis (70% to

99%), was treated with single antiplatelet therapy after 90 days of dual

antiplatelet therapy (aspirin and clopidogrel), and combined stent therapy was

not recommended. (Class III, Level of Evidence B).

[3] For stroke patients with intracranial arterial stenosis, aspirin plus clopidogrel

is recommended to reduce the risk of early stroke recurrence caused by

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 130

thromboembolism. One week later, the risk is reassessed for the purpose of

determining whether to continue the combined treatment. The duration of dual

antiplatelet therapy can last for 3 months after the onset of the disease. Routine

anticoagulant therapy is not recommended for secondary prevention (Class I,

Level of Evidence A).

(2) Surgical intervention: combined intracranial and extracranial macrovascular

stenosis and hemodynamic mechanism

1. Carotid endarterectomy The safety of emergency CEA is still uncertain. An

observational study included 369 stroke patients with CEA operation interval ≤ 1

week. The results suggested that early CEA operation after stroke was feasible and

relatively safe [519]. A study enrolled a total of 193 patients who underwent CEA due

to symptomatic stenosis, including 90 AIS patients and 27 TIA patients. The results

suggested that patients undergoing emergency CEA surgery had a higher risk than

selected patients undergoing CEA surgery [520]. Another study included 3023 patients

with carotid artery stenosis, 176 of whom underwent CEA within 48 hours due to

acute progressive stroke or transient ischemic attack. This study suggested that the

risk of CEA within 48 hours of onset for selected patients with progressive stroke or

TIA was within an acceptable range, and the benefit of CEA for symptomatic patients

at an early phase was to prevent recurrence of stroke [521].

Paty et al’s[522]study showed that for patients with infarct size larger than 1cm in

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 131

diameter, the risk of permanent neurological impairment after CEA increased by 1.7

times. Therefore, early CEA may be suitable for mild and non-disabling stroke, and

its purpose is to reduce continuous thromboembolism or flow-restrictive ischemia.

The results of this study also suggested that the incidence rate of postoperative stroke

deterioration and the postoperative results of CEA patients were similar and

acceptable within one month of stroke, and it is considered that surgery at any time

within one month after stroke onset is feasible.

Recommendations

[1] When clinical indicators or brain imaging demonstrate that the core of small

infarction and at risk area (penumbra) are caused by insufficient blood flow due

to severe stenosis or occlusion of the carotid artery, or acute neurological

impairment after CEA with suspected acute thrombosis at the site of operation,

the effectiveness of emergency CEA has not been confirmed (Class II, Level of

Evidence B).

[2] The effectiveness of emergency CEA has not been proven in patients with

unstable neurological status (such as progressive stroke) (Class II, Level of

Evidence B).

2. Other surgical treatment A study in 2013 included only 20 AIS patients due to

cerebrovascular atherosclerosis. Superficial temporal artery-middle cerebral artery

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 132

(STA-MCA) bypass surgery was given within 7 days after symptoms appeared.

Results suggested that early bypass surgery was safe and effective, and some of the

patients showed rapid improvement of neurological function. Therefore, some

selected AIS patients with small infarcts visible on imaging may benefit from early

STA-MCA bypass surgery [523].

Intracranial and extracranial vascular bypass surgery for the treatment of ischemic

stroke is considered ineffectual. Little literature has reported the benefits of early

bypass surgery, nor did they report haemorrhagic complications. Intravascular therapy

seems to be a better choice in most cases.

II. Management of cardiogenic stroke

(1) Anticoagulation treatment initiation

Cardiogenic stroke is the most common and serious type of stroke besides

atherosclerosis. For the treatment of cardiogenic stroke, besides actively treating

primary heart condition, anticoagulant therapy should be started based on the situation

to prevent stroke recurrence.

The study "Heparin in Acute Embolic Stroke Trial" (HAEST) is the only RCT study

to research the timing of anticoagulation. The results showed that patients without

high risk factors of haemorrhage had a low risk of haemorrhage when low molecular

weight heparin (LMWH) or aspirin was applied within 30 hours. The European Atrial

Fibrillation Trial (EAFT) showed that anticoagulation therapy was effective within 14

days after onset [524]. ACCP recommended in 2012 that anticoagulation therapy should

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 133

be started within 2 weeks for patients with non-large cerebral infarction and

cardiogenic embolism without other haemorrhagic risks. For patients with a high risk

of bleeding, large infarct area or poor blood pressure control, the anticoagulation time

should be extended to later than 14 days [567].The HAS-BLED score can be used to

predict bleeding risk of patients undergoing anticoagulant therapy, and patients with a

total score of ≥ 3 points are regarded as high-risk patients prone to haemorrhagic

transformation.

For new oral anticoagulants, their anticoagulant effects should not be inferior or

superior to warfarin, and their cerebral haemorrhage complications should be less

than that of warfarin, with high safety. Some foreign studies suggest that the size and

severity of infarct lesion should be taken into account when anticoagulation is

considered, suggesting that anticoagulation treatment can be initiated one day after

TIA. Non-disabling small-area infarction should be anticoagulated 3 days after onset

and moderate-area infarction should be anticoagulated 6 days after onset. However,

massive infarction should be anticoagulated at least 2-3 weeks after onset [525].

Atrial fibrillation is the most common cause of cardiogenic stroke. A multicentre

retrospective cohort study included 1,029 AIS patients newly diagnosed with atrial

fibrillation. Anticoagulant therapy was initiated within 4-14 days after stroke,

showing a better 90-day composite outcome, including stroke, TIA, systemic

embolism, symptomatic intracranial haemorrhage and severe extracranial

haemorrhage (comparation between anticoagulation initiated 4-14 days after stroke

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 134

and within 4 days, HR=0.53, 95% CI: 0.30~0.93). High CHADS2-VASC score, high

NIHSS score, large infarct size and anticoagulation type were associated with adverse

outcomes [526]. A retrospective, open-label study included 60 patients with atrial

fibrillation and mild to moderate AIS (NIHSS < 9), suggesting that patients treated

with rivaroxaban within 14 days after onset had no symptomatic haemorrhage within

7 days after anticoagulation was initiated [527].

Recommendations

[1] For patients with non-massive cerebral infarction and not from cardiogenic

embolism and without other bleeding risks, anticoagulant therapy is

recommended and to be initiated within 2 weeks (Class IIa, Level of Evidence B).

[2] For patients at high risk of bleeding, or large infarction area or poor blood

pressure control, the timing of initiation of anticoagulation therapy should be

extended to beyond 2 weeks (Class IIa, Level of Evidence B).

[3] The size and severity of stroke should be taken into account when consider

anticoagulation. It is suggested that anticoagulation can be initiated 1 day after

TIA, 3 days after non-disabling small infarction, and 6 days after moderate size

infarction. Large area infarction should wait at least 2 to 3 weeks (Class IIa,

Level of Evidence B).

[4] For most AIS patients with atrial fibrillation, it is reasonable to start oral

anticoagulant therapy within 4 to 14 days after onset (Class IIa, Level of

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 135

Evidence B).

(II) Drug selection

1. Oral anticoagulant Warfarin has definite therapeutic value in primary

prevention[528] and secondary prevention[524] of stroke in patients with atrial

fibrillation. The best dosage of warfarin anticoagulant therapy is when INR value is of

2.0-3.0, which gives due consideration to the curative effect and bleeding risk [529].

For patients with atrial fibrillation who still suffer from ischemic stroke or TIA after

anticoagulant therapy, there is no evidence to support the prevention of ischemic

events by increasing drug dosage.

The new oral anticoagulants are convenient to take, do not require adjustment of the

dosage or frequent monitoring of the INR value, and have clear benefits and low

bleeding risks for patients with non-valvular atrial fibrillation. These anticoagulants

have been recommended by the guidelines of various countries in recent years.

Several RCT studies have verified the efficacy and safety of , rivaroxaban,

and in preventing stroke and embolism events in patients with

atrial fibrillation [530-534]. New oral anticoagulants provide a new choice for prevention

of thromboembolic complications in patients with atrial fibrillation. However, due to

limited application time and clinical experience in China, it is still difficult to be

widely used, therefore warfarin is still the preferred oral anticoagulant. The 2016

Canadian Guidelines for Atrial Fibrillation Management emphasizes that warfarin,

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 136

instead of new oral anticoagulant drugs, should be the first choice for patients with

mechanical valves, rheumatic mitral stenosis and glomerular filtration rate of 15-30

ml/(min·1.73m2) with indications of oral anticoagulants [567].

Recommendations

[1] For ischemic stroke or TIA patients with atrial fibrillation (including

paroxysmal), appropriate doses of warfarin are recommended to prevent the

recurrence of thromboembolism. The target dose of warfarin is to maintain INR

at 2.0 to 3.0 (Class I, Level of Evidence A).

[2] New oral anticoagulants can be used as an alternative to warfarin. New oral

anticoagulants include dabigatran, rivaroxaban, apixaban and edoxaban (Class

I, Level of Evidence A). Individual factors should be taken into account in the

selection of drugs.

2. Heparin AIS has been treated by intravenous application of anticoagulants for

more than 50 years, but this have become less common. Reasons to use such

anticoagulants for acute stroke include: ① prevent aggravation of neural function; ②

prevent early embolism recurrence; and ③ improve neural outcome[535-536] The

previous AHA expert consensus considers that the results of safety and effectiveness

of heparin or other anticoagulants administrated during acute phase are negative or

uncertain [537-539]. Other investigators also believe that the available clinical trial data

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 137

cannot support the effectiveness of emergency anticoagulation in the treatment of

recurrent ischemic stroke [540-541]. Two Meta analyses also verify that the emergency

anticoagulation is lack of benefit [542-543]. Another non-blind RCT study not included

in the two Meta analyses compared the efficacy of low molecular heparin (LMWH)

and aspirin on prevention of early neural function aggravation. Though LMWH was

superior than aspirin in subgroups based on age (LMWH, 63.82% vs Aspirin 44.63%;

P<0.001) and posterior infarction (LMWH, 75.19% vs Aspirin 40.48%, 0<0.001), the

difference was not significant for subgroups analysis of aaetiology, sex, NIHSS score,

and anterior infarction [544].

In a random, double-blind placebo control test using intravenously, the

subgroup analysis for different pre-set subtypes of patients showed that only the

patients with stenosis > 50% due to aortic atherosclerosis benefited from the

treatment, specifically, 53.8% of patients in the danaparoid group compared to 38.0%

of patients in the placebo group achieved good outcome at 7d (P = 0.023) [545]. The

result is consistent with the result of early recurrence rate of stroke patients due to

serious atherosclerosis found in other studies [546]. A random test conducted in

Singapore and Hong Kong compared Asian patients taking aspirin or nadroparin

within 48h of onset for aortic atherosclerotic stroke. Almost all patients had serious

stenosis or intracranial artery occlusion, and there were a few patients with

extracranial arterial disease. The test showed that there was no significant difference

in either haemorrhage or clinical benefit [547]. A multicentre study discussed the

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 138

efficacy of subcutaneous injection of enoxaparin or dose-adjusted unfractionated

heparin in the highly stenosis or cardioembolic stroke patients, and no significant

difference was found [548].

The optimal pharmacotherapy is still uncertain for AIS patients with imaging

evidence of nonocclusive intraluminal thrombosis (such as carotid artery and

vertebrobasilar artery). Several small observation studies confirmed the safety of

short-term intravenous infusion of heparin or LMWH in such population [549-550].

More studies are needed to be verified of its effectiveness and safety.

Recommendations

[1] Emergency anticoagulant therapy for AIS patients is not recommended for

the prevention of early recurrence of stroke, stop the deterioration of

neurological function and improve the outcome of AIS (Class III, Level of

Evidence A).

[2] The effectiveness of emergency anticoagulant therapy is unclear in AIS

patients with severe ipsilateral internal carotid artery stenosis (Class IIb, Level

of Evidence A).

[3] For AIS patients with extracranial intravascular non-occlusive thrombosis,

the safety and efficacy of short-term anticoagulant therapy are unclear (Class

IIb, Level of Evidence C).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 139

(III) Aaetiology management

1. Atrial fibrillation One of the important complications of atrial fibrillation is

cardiogenic brain embolism. The studies show that oral administration of warfarin can

effectively prevent ischemic stroke [567] in atrial fibrillation patients, and the risk of

stroke is reduced by more than 60% [583]. Therefore, theroretically, in absence of

contraindications, all atrial fibrillation patients having had stroke attack shall take

anticoagulants for a long term. However, in the clinical practice, warfarin is

desperately underused by atrial fibrillation patients [567], and the treatment rate with

warfarin for ischemic stroke patients with atrial fibrillation in China is only 16.2%

[528]. A meta-analysis shows aspirin alone is effective for ischemic stroke or TIA patients

with atrial fibrillation if failing to take anticoagulants orally [551]. Atrial Fibrillation

Clopidogrel Trial With Irbesartan for Prevention of Vascular Events (ACTIVE-A)

verified the benefits of a combination of aspirin and clopidogrel in atrial fibrillation

patients non-applicable for anticoagulation, however with increased risk of

haemorrhage[552]. The ACTIVE-W study confirmed the advantage of anticoagulation

over dual antiplatelet therapy in atrial fibrillation patients [553]. The EAFT study also

confirmed the advantage of anticoagulation over dual antiplatelet therapy in TIA or

mild stroke patients with atrial fibrillation [524].

In China, the incidence of atrial fibrillation is 11.45% in the initial ischemic stroke or

TIA patients, which is significantly lower than the incidence in foreign countries

(17.8-24.6%), showing low detection rate of atrial fibrillation in the ischemic stroke

or TIA patients in China. Newly developed atrial fibrillation is detected in about 10%

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 140

of ischemic stroke or TIA patients during admission by the current conventional

examination means (routine ECG or 24h ECG). The Stroke and Monitoring for PAF

in Real Time (SMART) study adopted a consecutive 30d ECG monitoring, which can

increase the detection rate of atrial fibrillation by 11% [554]. Event Monitor Belt for

Recording Atrial Fibrillation after a Cerebral Ischemic Event (EMBRACE) included

cryptogenic ischemic stroke or TIA patients without atrial fibrillation as verified by

routine 24h dynamic ECG monitoring as the study population, compared the detection

rates of paroxysmal atrial fibrillation between 30d ECG monitoring and repeated 24h

dynamic ECG monitoring, and showed that only 4% of atrial fibrillation was found in

the repeated 24h dynamic ECG monitoring records, 20% in 30d ECG records. The

detection rate of atrial fibrillation may be increased by prolonging the ECG

monitoring time, which is of great importance for the cardiogenic embolism caused

by atrial fibrillation [555].

Recommendations

[1] For ischemic stroke or TIA patients with atrial fibrillation, the time of

anticoagulation should be chosen according to the severity of ischemia and the

risk of bleeding transformation. It is suggested that anticoagulation therapy

should be given within 14 days after the onset of neurological symptoms to

prevent stroke recurrence. For patients with high risk of bleeding, the timing of

anticoagulation should be appropriately prolonged (Class IIa, Level of Evidence

B).

[2] If ischemic stroke or TIA patients with atrial fibrillation are unable to receive

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 141

oral anticoagulant therapy, aspirin alone may be considered for treatment (class

IIa recommended, class B evidence). Aspirin combined with clopidogrel should

be carefully selected and antiplatelet therapy (Class IIb, Level of Evidence B).

2. Other cardiogenic embolisms Ischemic stroke following acute myocardial

infarction is one of the exocardial complications of myocardial infarction. Left

ventricular mural thrombosis easily occurs due to massive myocardial infarction,

particularly in anterior myocardial infarction with apex involvement. Anticoagulation

should be taken into consideration to prevent thrombosis development if the patient

has low haemorrhagic risks. Mural thrombosis, once diagnosed, shall be treated with

oral administration of . If stenting and dual antiplatelet therapy

have been performed, the addition of oral administration of anticoagulants may

increase the haemorrhagic risks of patients. Therefore, anticoagulation plus dual

antiplatelet therapy may only be used for ST-segment elevation myocardial infarction

patients whose risks of systemic circulation embolism or venous thromboembolism

are higher than the haemorrhagic risks. When triple antithrombotic therapy is needed,

the range of INR should be controlled to be 2.0-2.5. Valvular heart diseases (mitral stenosis, mitral annulus calcification, mitral

regurgitation, mitral prolapse, aortic valve disease, heart valve prosthesis and

biovalve) may also increase the risk of cerebrovascular disease events caused by

cardiogenic embolism. Antithrombotic therapy for valvular heart diseases is of great

significance for reducing thrombosis. However, the possible increase of haemorrhagic

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 142

risk must be taken into account. Therefore, antithrombotic therapy should be used to

maintain the best balance between thrombosis and haemorrhagic risk [567]. In

summary, patients with cardiogenic embolic stroke due to heart diseases shall seek

medical advices in the cardiology department as early as possible.

The incidence of patent foramen ovale is 15-25% in adults, and the incidence of atrial

septal aneurysm is 1-4%. A right-to-left shunt channel may be formed due to patent

foramen ovale, resulting in paradoxical embolism from the venous system, while

atrial septal aneurysms easily result in thrombosis. Patent foramen ovale and atrial

septal aneurysms are associated with the stroke in many but not all studies [556-564]. In

the PICSS study, it was shown through transoesophageal ultrasound scan that the

incidence of patent foramen ovale was higher in the patients with cryptogenic stroke

than in patients with stroke due to known reasons (39.2% vs. 29.9%)[557, 565] However,

not all of the stroke patients with patent foramen ovale have paradoxical embolism

from deep venous system. It’s suggested that the cryptogenic stroke patients with

patent foramen ovale should undergo Doppler ultrasound scan for veins in lower

limbs to exclude the deep venous thrombosis. The incidence of deep venous

thrombosis in stroke patients is about 7.6%. Routine pelvic MR venography remains

to be discussed for screening of cryptogenic stroke [565].

RESPECT study included 980 PFO patients, with median follow-up time of 5.9 years.

RESPECT study showed that the risk of stroke recurrence after PFO occlusion by

Amplatzer PFO occluder (Abbott vascular, former St.Jude medical) was reduced[566].

The REDUCE study was one of the first studies to use Helex (Gore&Associates) and

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 143

Cardioform ventricular septal occluders (Gore&Associates). It compared patients

undergoing closure therapy and long-term antiplatelet therapy with the patients

undergoing antiplatelet therapy alone. REDUCE study included 664 patients, median

follow-up time was 3.2 years. Results indicated PFO occlusion patients had obvious

clinical benefits [567]. The two studies above show that PFO occlusion therapy indeed

reduces the risk of recurrent ischemic stroke, and that PFO occlusion related risk is

very low.

Recommendations

[1] In ischemic stroke or TIA patients with acute myocardial infarction, and left

ventricular mural thrombus, warfarin oral anticoagulation therapy is

recommended for at least 3 months (target INR = 2.5, range 2.0 to 3.0) (Class IIa,

Level of Evidence B).

[2] If there is no left ventricular mural thrombus, but no movement or abnormal

movement of the anterior wall is found, oral anticoagulation therapy of warfarin

for 3 months should also be considered (target INR value = 2.5, range 2.0 to 3.0)

(Class IIa, Level of Evidence B).

[3] For ischemic stroke or TIA patients with rheumatic mitral valve disease but

without atrial fibrillation and other risk factors, such as carotid stenosis, oral

anticoagulation therapy with warfarin is recommended (target INR = 2.5, range

2.0 to 3.0) (Class IIa, Level of Evidence B).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 144

[4] Patients with rheumatic mitral valve disease who have been treated with

warfarin, routinely combining with antiplatelet therapy after ischemic stroke or

TIA is not recommended (class III recommended, class C evidence). However,

aspirin antiplatelet therapy can be added when ischemic stroke or TIA still

occurs during the treatment of sufficient amount of warfarin (Class IIa, Level of

Evidence B).

[5] Ischemic stroke or TIA patients with non-rheumatic mitral valve disease or

other valve diseases (local aortic arch, mitral annulus calcification, mitral valve

prolapse, etc.) without atrial fibrillation may consider antiplatelet therapy (Class

IIa, Level of Evidence B).

[6] For patients with ischemic stroke or TIA and mechanical artificial heart

valve, long-term warfarin oral anticoagulation therapy is recommended (INR

2/5-3.5) (Class IIa, Level of Evidence B).

[7] For patients with previous history of ischemic stroke or TIA and mechanical

artificial heart valves, if the risk of bleeding is low, aspirin can be used in

addition to warfarin anticoagulation (Class IIa, Level of Evidence B).

[8] It is suggested that any decision on PFO occlusion should be made jointly by

neurologists and cardiologists (Class I, Level of Evidence A).

[9] Before PFO occlusion, other known causes of ischemic stroke (including

monitoring arrhythmias) should be carefully excluded. The possibility of PFO

correlation with the stroke, risk factors, and lifestyle changes should be assessed.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 145

And communication between patients and multidisciplinary clinical teams

should be involved in making the decision. For ischemic stroke caused by PFO,

PFO occlusion can be performed to reduce the risk of stroke recurrence (Class I,

Level of Evidence A).

III. Small vessel disease

Lacunar infarction caused by cerebral small vessel disease accounts for 25-50% of

ischemic stroke, while the recurrence rate of stroke caused by small vessel disease is

slightly lower than that of stroke caused by great vessel atherosclerosis. Cerebral

arteriolar hyaline degeneration or amyloidosis has different pathological changes

compared to atherosclerosis, and antithrombotic therapy on this population may have

poorer efficacy than that of large arterial stroke. A single antiplatelet agent, including

aspirin, clopidogrel, and cilostazol, should be administrated as secondary prevention

for newly-developed symptomatic subcortical small infarctions. Several studies show

that long-term administration of a combination of two antiplatelet agents will increase

the risk of cerebral haemorrhage, which causes the disadvantages to outweigh

advantages. SPS3 study showed dual use of aspirin and clopidogrel could not reduce

the recurrence risk of stroke, but increased the risk of cerebral haemorrhage as

compared with aspirin alone[140]. Many patients with symptomatic subcortical small

infarction may have concurrent multiple lacunar infarction, white matter

hyperintensities and microbleeding, increasing the risk of haemorrhage. For those

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 146

patients, cilostazol can be chosen if antiplatelet therapy is needed.

The occurrence and development of age and vascular risk factor related small vessel

disease are very closely associated with hypertension. Elevated systolic and diastolic

arterial pressure is an independent risk factor for the occurrence and development of

cerebral small vessel disease. However, it’s not enough to control the systolic pressure

and diastolic pressure in normal ranges. High fluctuation of blood pressure will also

accelerate development of the small vessel disease. The hypertensive cerebral

haemorrhage is usually accompanied with drastic fluctuation of blood pressure before

attack. Too high blood pressure variability will promote the progress of cerebral small

vessel disease. The standard deviation of change in systolic or diastolic pressure,

coefficient of variation and mean-independent variation may be used as parameters to

evaluate the variability of blood pressure. Ideal blood pressure fluctuation range is yet

unknown. However, we should pay attention to blood pressure variability during

visits in addition to control of systolic and diastolic pressure. Increases blood pressure

variability is an independent risk factor of development of cerebral microbleeding.

Too high or too low blood vessel change will aggravate the clinical symptoms of

cerebral small vessel disease, causing dizziness, instability of gait or vascular

cognitive function decline, and even result in cerebral haemorrhage or lacunar

infarction.

Recommendations

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 147

[1] The mechanism of ischemic stroke caused by small vascular disease is

complex. At present, it is recommended to manage blood pressure, and use of

aspirin, or clopidogrel or cilostazol (Class I, Level of Evidence B).

[2] Cerebral small vessel disease leads to a significant decrease in the

adaptability of brain tissue to the changes of excessive hypertension and

hypotension. The blood pressure of patients should be closely monitored (Class

IIa, Level of Evidence B).

[3] Control of systolic and diastolic pressure is the key factor to control the

incidence and progression of cerebral small vessel disease (Class IIa, Level of

Evidence B).

[4] It is necessary to monitor the 24-hour ambulatory blood pressure in patients

with cerebral small vessel disease. When conditions permit, it is best to detect

changes in blood pressure during head upright tilt test (Class I, Level of

Evidence B).

IV. Management of stroke due to special aaetiology

(I) Aortic dissection

CADISS study team published a random, open-label, phase Ⅱ anticoagulation vs.

antiplatelet feasibility trial, which enrolled 250 patients with extracranial carotid

artery or vertebral artery dissection from 46 centres in UK and Australia[568]. The

primary outcome was ipsilateral stroke or all-cause death 3 months after

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 148

randomization by intention to treat analysis, and there was no significant difference

between both groups. There was also no difference in serious haemorrhage rate. The

event incidences of both groups were low in the phase Ⅱ clinical trial, therefore the

phase Ⅲ clinical trial might not be carried out. The limitations of the study also

include lack of central imaging data in 20% of cases, and mean randomization time of

3.65 days, which is not enough to cover the hyperacute cases. Nonetheless, CADISS

study confirmed the conclusion of many previous observational studies, i.e., no

significant difference between the carotid artery dissection patients treated by

anticoagulation and antiplatelet therapy. In addition, the follow-up analysis found no

difference in the natural history of dissecting aneurysm and relevant stroke risk

between two treatment groups, showing the overall prognosis was good [569].

(II) Moyamoya disease and moyamoya syndrome

Moyamoya disease is a cerebrovascular disease featured by chronic progressive

stenosis or occlusion of bilateral terminal internal carotid arteries, anterior cerebral

artery or middle cerebral artery, with secondary abnormal vessel network. Moyamoya

disease and moyamoya syndrome have complex and various clinical manifestations.

Cerebral ischemia is most common, manifested as transient ischemic attack (TIA),

reversible ischemic neurologic deficit (RIND) or cerebral infarction. TIA is usually

induced by emotional stress, crying, strenuous exercise or eating of hot spicy food.

For moyamoya disease, angiography is a gold standard for diagnosis. HR-MRI is also

of some help to the diagnosis of moyamoya disease. Willis circle proximal aorta

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 149

involvement is most bilateral; decrease of external diameters of vessels, generally

without positive remodelling; obvious vascular wall thickening, obvious luminal

stenosis; mild concentric strengthening, irrelevant to symptoms, generally no

inflammatory cell infiltration on pathological examination. There’s no definitely

effective drug for moyamoya disease at present. A major therapy for moyamoya

disease and moyamoya syndrome is extracranial-intracranial vascular reconstruction,

which can effectively prevent and treat ischemic stroke.

(III) Vasculitis due to various reasons

Its incidence is low at only 2.4/1 million persons·year. It is classified as primary

vasculitis and secondary vasculitis (autoimmune/infectious), and should be diagnosed

and treated early. Diagnosis criterion of primary central nervous system vasculitis:

acquired or other unexplainable neural or mental abnormality; vasculitis verified by

angiography or tissue biopsy; no other evidence of secondary vasculitis. Brain tissue

biopsy has low sensitivity (less than 50%), and is invasive, while the angiography (for

luminal stenosis, tumour-like dilation and beaded change) has low specificity.

On HR-MRI, vasculitis has the following imaging characteristics: vascular

distribution - intracranial artery and arteriole with diameter of 100-500μm,

intracranial proximal aorta and multiple vessels can also be involved; vessel wall

thickening - mainly concentric thickening, less eccentric thickening, the same signal

as grey matter on T2WI; vascular wall strengthening - uniform, smooth and

concentric, associated with inflammation activity level, strengthening degree may be

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 150

reduced by hormonotherapy; vascular wall strengthening may vary during follow-up,

but luminal stenosis generally has little change, and its recovery period is long.

Thickening and strengthening of vascular wall of vasculitis patient; sustained stenosis

may be seen after follow-up for 3 months. If diagnosed with central nervous system

vasculitis, the patients shall receive hormonotherapy or immunosuppressor treatment.

(IV) Cerebral venous system disease

Venous infarction is derived from venous sinus thrombosis, cortical venous

thrombosis and deep venous thrombosis. Venous infarction is mainly featured by

noncompliance with arterial blood supply distribution region and high probability of

haemorrhage, mainly vasogenic ooedema (as compared to cytotoxic ooedema). High

density is shown on CT for small venous thrombosis. MR manifestations depend on

the blood composition, but there should be no flow void effect in any case. Deep

venous thrombosis is usually manifested as unilateral or bilateral (more common)

thalamic oedema (thrombosis in internal cerebral vein). Venous infarction is mainly

featured by severe cerebral oedema and haemorrhage. Sometimes it causes idiopathic

intracranial hypertension and does not result in brain parenchyma injury. HR-MRI can

be also used for evaluating the intracranial venous conditions of such patients.

Recommendations

[1] For ischemic stroke patients with defined aetiology, targeted etiological

treatment is needed (Class I, Level of Evidence A).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 151

[2] For AIS patients with extracranial carotid or vertebral artery dissection,

antiplatelet or anticoagulant therapy for 3 to 6 months may be reasonable (Class

IIa, Level of Evidence B).

[3] For patients with moyamoya disease and smoke syndrome, it is suggested that

active drug treatment should be taken for underlying diseases or complications,

and the risk factors of stroke should be effectively controlled and managed. It is

necessary to choose the appropriate time and mode of operation according to the

evaluation of patients (Class IIa, Level of Evidence B).

[4] The diagnosis of vascular inflammatory diseases in the central nervous

system is difficult, and the etiological treatment should be carried out on the

basis of definite diagnosis (Class IIa, Level of Evidence B).

[5] For patients with suspected venous cerebral infarction, it is suggested to

complete intracranial venous system angiography. After that, according to the

clinical and imaging results, these patients will receive etiological and

symptomatic treatment (Class IIa, Level of Evidence B).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 152

Section 9 Management of risk factors and long-term intervention

I. Blood pressure management

Hypertension is the most important risk factor of stroke and TIA. The diagnosis rate

of hypertension in the patients with the attack of ischemic stroke recently is as high as

70% [329, 596]. The blood pressure management post stroke is an issue of concern.

There are many disputes on the initiation opportunity of antihypertensive therapy and

target blood pressure.

(I) When to initiate antihypertensive therapy

Several clinical random control studies including PRoFESS, ACCESS and SCAT

studies, and meta-analysis consistently show it’s safe to initiate the antihypertensive

therapy within 48-72h after the occurrence of an AIS event, but the improvement of

functional prognosis or decrease of mortality is not definite [570]. An ongoing China

Antihypertensive Trial in Acute Ischemic Stroke II (CATIS-2) initiated in 2017 will

explore the efficacy of antihypertensive therapy within 24-48h after AIS, and is

intended to test if the risk of death and serious disability can be reduced by early

antihypertensive therapy within 24-48h after AIS as compared to delayed

antihypertensive therapy (7d after attack).

Another question as to when to initiate the antihypertensive therapy is: whether

hypertensive patients previously taking anti-hypertensive drugs shall stop or continue

taking anti-hypertensive drugs in the acute phase of ischemic stroke? COSSACS and

ENOS studies answer this question. Both of these trials point out that the patients

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 153

cannot benefit from the administration of hypotensive drugs in the acute phase of

ischemic stroke, but it does not result in aggravation or poor prognosis [571]. China

Guideline for Secondary Prevention of Ischemic Stroke and Transient Cerebral

Ischemic Attack 2014 points out that the antihypertensive therapy shall be restarted

several days after attack for the ischemic stroke or TIA patients having hypertension

history and taking hypotensive drugs for a long term in case of no absolute

contraindications [572].

A large sample size random prospective study showed the antihypertensive therapy

within 24 could not improve the 14d disability rate or death rate [573]. Later, the study

found antihypertensive therapy initiated within 24-48h after the ischemia event could

improve the clinical prognosis at 3 months and reduce the recurrence and death rates

[574]. In addition, a large sample size random control study with follow-up for 6

months showed that the higher baseline blood pressure or high blood pressure

variation within 24h was associated with poor prognosis, but the prognosis could be

improved by antihypertensive therapy within 24h [575]. Patients under thrombolysis

have better prognosis if their blood pressure is controlled < 160mmHg as compared

with the patients whose blood pressure is > 160mmHg, the patients whose blood

pressure is controlled < 140mmHg have better prognosis than above mentioned

patients[576], and many studies support that the patients may benefit from the early

blood pressure control (140-160)/(80-99)mmHg after stroke [577-578].

However, a study and 2 meta analyses show that the hypotensive drugs can effectively

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 154

lower the blood pressure at the acute phase, but have no influence on the short-term

and long-term prognosis and death rate [579-583]. Relatively small sample size random

control studies also find that antihypertensive therapy has no obvious influence on the

prognosis [584], but may increase the risk of early neurological deterioration [471-472, 585-

586]. A study shows that the antihypertensive therapy possibly has different influence

on prognosis according to different types of stroke [587].

(II) Management target of blood pressure during acute phase

1. For the general population The China Antihypertensive Trial in Acute Ischemic

Stroke (CATIS) observed 4071 ischemic stroke patients at acute phase (within 24h

after admission) within 48h after ischemic attack. The influence of intensified

antihypertensive therapy on death and serious disability at 14d, during discharge and

at 3 months in the patients whose blood pressure was above 140/90mmHg was

analysed, showing that the patients in the intensified antihypertensive therapy group

did not obviously benefit from it, but it might be safe to control the blood pressure

below 140/90mmHg [557].

There’s no definite study conclusion on the control target of blood pressure from

which the patients may get maximum benefits after AIS. Some observation studies

confirmed that lower blood pressure was associated with poor prognosis after stroke,

but this conclusion is disputed in different studies [473, 588-594]. There’s no study on the

hypotension treatment for stroke patients. A meta-analysis including 12 studies

compared the influence of crystalloid solution and colloidal solution on prognosis,

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 155

showing that the death rates and disability rates were similar. There’s no definite

study data which may be used to instruct the dose and application duration of such

expansion solutions [595]. There’s no study comparing the expansion effects of

different isotonic fluids. There’s no definite study result on the treatment significance

of drug-induced hypertension among AIS patients.

Recommendations

[1] For patients with blood pressure < 220/120 mm Hg, who do not receive IV rt-

PA or endovascular treatment and do not have complications requiring

emergency antihypertensive treatment, starting or restarting antihypertensive

therapy within the first 48 to 72 hours after AIS is not effective in preventing

death or severe disability (Class III, Level of Evidence A).

[2] For patients with blood pressure ≥ 220/120 mm Hg, who do not receive IV rt-

PA or endovascular treatment and do not have complications requiring

emergency antihypertensive treatment, the effect of starting or restarting

antihypertensive therapy within the first 48 to 72 hours after AIS is uncertain. It

may be reasonable to reduce blood pressure by 15% within the first 24 hours

after a stroke attack (Class IIb, Level of Evidence C).

2. Special populations (Patients with ICAS, medical complications, and blood

pressure > 220/110mmHg) AIS patients may have other serious comorbidities, and

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 156

emergency antihypertensive treatment needs to be initiated to prevent more serious

conditions. However, in some cases, excessively rapid blood pressure reduction may

lead to aggravation of intracranial ischemia [505], so antihypertensive treatment needs

to be vigilant. In this case, the ideal antihypertensive treatment should be to achieve

an overall antihypertensive effect of 15% through individualized treatment.

Patients with very high blood pressure (blood pressure > 220/120mmHg in general)

have been excluded in existing studies [557, 596-600]. Therefore, for these patients, the

antihypertensive effect has not been clearly demonstrated in the absence of a clear

medical complication.

Recommendations

[1] For AIS patients with other complications (such as simultaneous acute

coronary events, acute heart failure, aortic dissection, bleeding transformation

after thrombolysis, or preeclampsia/eclampsia), early antihypertensive therapy is

indicated. At the initial stage, a 15% reduction in blood pressure may be safe

(Class I, Level of Evidence C).

[2] Hypotension and hypovolemia must be corrected after stroke to ensure

systemic perfusion to support organ function (Class I, Level of Evidence C).

[3] For AIS patients, the therapeutic effect of drug-induced hypertension is

uncertain (Class IIb, Level of Evidence C).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 157

(III) Antihypertensive target level in secondary prevention of hypertension

In the Post-Stroke Antihypertensive Treatment Study (PATS) conducted in China to

investigate the effectiveness of antihypertensive treatment in secondary prevention of

hypertension, 5,665 patients with recent TIA or minor strokes (haemorrhagic and

ischemic) were selected and randomly divided into indapamide treatment group and

placebo group. The results after an average follow-up time of 24 months indicated

that the recurrence rate of stroke in the indapamide group was significantly lower than

that in the placebo group (30.9% vs. 44.1%), and the relative risk of stroke recurrence

was reduced by 30% [601]. The early Perindopril Protection Against Recurrent Stroke

Study (PROGRESS) has once again confirmed the effectiveness of blood pressure

control in secondary prevention of stroke [602]. A meta-analysis conducted in 2009 has

demonstrated that antihypertensive treatment can significantly reduce the recurrence

risk of stroke and TIA, and the greater the reduction in systolic blood pressure, the

more significant the effect of reducing the risk of stroke recurrence [603].

Both clinical trials have demonstrated that initiating or continuing antihypertensive

medication after hospitalization can improve the blood pressure management [604, 605].

For hypertension patients with stable neurological function during hospitalization, it is

reasonable to initiate or restart antihypertensive treatment. These studies have

evaluated patients with a previous history of hypertension. Since hypertension is

commonly detected and diagnosed for the first time after admission to the hospital

due to stroke, the above findings are still applicable to patients without a previous

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 158

history of hypertension. For patients with ischemic stroke or TIA induced by different

causes, there is still no basis for determining the target value of hypotension. For

patients with ischemic stroke or TIA due to atherosclerotic stenosis of intracranial

artery (stenosis rate of 70% to 99%), systolic blood pressure is recommended to be

reduced to less than 140 mmHg and the diastolic blood pressure to be reduced to 90

mmHg [606]. For patients with stroke or TIA due to low hemodynamic factors, the

effect of blood pressure lowering velocity and amplitude on patients’ tolerance and

hemodynamic parameters should be weighed [607]. A total of 3,020 patients with

lacunar infarction were included in the Secondary Prevention of Small Subcortical

Strokes (SPS3) study, and randomly (non-blind) divided into two groups (Target

systolic pressure < 130mmHg vs. 130-149 mmHg). Although there was no

statistically significant difference in the risk of stroke recurrence between the two

groups, the proportion of patients with cerebral haemorrhage in the group with

systolic blood pressure < 130mmHg was drastically reduced, and the difference in the

proportion of severe hypotension between the two groups was not statistically

significant [608], which indicated that it may be more appropriate to control systolic

blood pressure at < 130mmHg for patients with subcortical infarction that may be the

cause of small vessel disease.

In the Systolic Blood Pressure Intervention Trial (SPRINT), the effects of intensive

antihypertensive treatment (target value < 120mmHg) and standard antihypertensive

treatment (< 140mmHg) on the risk of death and cardiovascular events were

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 159

compared [609]. The results indicated that the mean systolic blood pressure of patients

in the intensive antihypertensive treatment group and the standard antihypertensive

treatment group was 121.4 mmHg and 136.2 mmHg at 1 year, respectively. The

annual rate of major composite endpoint events in the intensive antihypertensive

treatment group was significantly lower than that in the standard antihypertensive

treatment group (1.65% vs. 2.19%, P< 0.001). All-cause mortality was also

significantly reduced in the intensive antihypertensive treatment group (risk

ratio=0.73, P=0.003). However, the incidences of severe hypotensive adverse events,

syncope, electrolyte abnormality, acute kidney injury or failure in the intensive

antihypertensive treatment group were higher than those in the standard

antihypertensive treatment group. Nevertheless, patients with diabetes, massive

proteinuria, history of stroke, end-stage renal disease, recent acute coronary syndrome

or patients hospitalized for heart failure, and other high-risk patients were all excluded

from this study, so the conclusions cannot be simply generalized to all patients with

hypertension.

The ACCORD study (Action to Control Cardiovascular Risk in Diabetes) in diabetic

patients has found that strict antihypertensive treatment strategies cannot reduce

patients’ primary endpoint events but can significantly reduce the risk of stroke events

[610]. The study believes that for diabetes patients, controlling systolic blood pressure

to less than 130/80 mmHg may benefit patients more, but the conclusions of the study

on patients with coronary heart disease, stroke, and chronic kidney disease are

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 160

unknown.

The ongoing Intensive Blood Pressure Intervention in Stroke Trial Sprint (IBIS) and

China Antihypertensive Trial in Acute Ischemic Stroke II (CATIS-2) are conducted

for further research on the target value of antihypertensive treatment in stroke

patients.

Recommendations

If the patient has stable neurological function during hospitalization, but blood

pressure > 140/90mmHg, it is safe to start or restart antihypertensive therapy.

With the exception of contraindications, long-term control of blood pressure is

reasonable (Class IIa, Level of Evidence B).

(IV) Selection of antihypertensive drugs

The benefit of antihypertensive treatment to reduce the risk of stroke mainly comes

from antihypertension itself. Various commonly used antihypertensive drugs can be

used as treatment options to control blood pressure in stroke patients. The RCT

research evidence in the field of stroke should be combined with the pharmacological

characteristics of different antihypertensive drugs and the individual situation of

patients for appropriately chosen antihypertensive drugs. There is no precise data on

the selection and dosage recommendation of antihypertensive drugs.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 161

Recommendation

Although there is not sufficient data to guide the selection of antihypertensive

drugs after AIS, the antihypertensive drugs and doses of figure 9 are reasonable.

(Class IIa, Level of Evidence C).

II. Management of abnormal lipid metabolism

The study of Stroke Prevention by Aggressive Reduction in Cholesterol Levels

(SPARCL) has shown that cholesterol level is one of the important factors for

recurrence of ischemic stroke or transient ischemic attack (TIA). Cholesterol level

lowering is important to reduce the recurrence of ischemic cerebrovascular disease

and death. Intensive cholesterol-lowering regimen (80 mg of atorvastatin daily) can

reduce the relative risk of stroke by 16% over 5 years [611]. Statins have a clear

secondary preventive effect on stroke and have a certain effect in improving stroke

prognosis.

(I) Time to start cholesterol-lowering treatment

In cases where blood cholesterol is not measured, statins may be recommended for

patients with stroke which is presumed to be caused by atherosclerosis [660].

Measurement of blood cholesterol may be valuable for ischemic strokes that are

presumed to be non-atherosclerotic, such as those caused by arterial dissection,

because primary prevention guidelines for such strokes are made based on LDL-C

levels [612].

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 162

There are only limited randomized controlled trials about early application of statins

after AIS. In the study on rapid assessment of stroke and transient ischemic attack to

prevent early recurrence (FASTER), the effects of 40mg of simvastatin and placebo in

the treatment of transient cerebral ischemia or mild stroke within 24h after attack

were evaluated [661]. The trial was terminated early because of slow enrolment. There

were no significant differences in recurrent stroke or safety endpoints between the

simvastatin group and placebo group. The effectiveness of the FASTER study was

insufficient due to early termination. The dose of statin used in this study was

moderate (not high-intensity statins as recommended in secondary stroke prevention).

When the application of statins started within 24 hours or within 7 days after attack,

there was no difference in outcome after 90s. The ASSORT study (a statin study after

AIS hospitalization) showed no difference in 90-day modified Rankin Scale (mRS)

between the group initiating statins within 24 hours and the group initiating statins

within 7 days [613].

Recommendations

[1] Routine measurement of blood cholesterol levels is not recommended for all

patients with atherosclerotic ischemic stroke who are not on high intensity statins

(Class III, Level of Evidence B).

[2] For patients with ischemic stroke during statins, it is reasonable to continue

statins in the acute phase of stroke (Class IIa, Level of Evidence B).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 163

[3] For patients who meet the requirements for statins, it is reasonable to start

statins during the hospital stay (Class IIa, Level of Evidence C).

(II) Low density lipoprotein (LDL) target levels of cholesterol-lowering

treatment after ischemic stroke

When the guideline development team reviewed randomized controlled trials on

cholesterol management related to cerebrovascular disease and cardiovascular

disease, no related records on the absolute target for low-density lipoprotein

cholesterol (LDL-C) of 1.8mmol/L (or 70mg/dl) were found, but the target value of

2.6mmol/L or 1.8mmol/L was adopted in multiple observational studies or guidelines

for reducing LDL-C [614-615]. The 2012 Canadian Cardiovascular Association

Guidelines recommend that the lipid-lowering target for patients with cerebrovascular

disease is set as LDL-C<2.0mmol/L or reduction of > 50% compared with baseline

[616]. For high-intensity statin therapy, it is still recommended to use LDL-

C<1.8mmol/L (70mg/dl) as the reference target value for cholesterol-lowering

treatment in consideration of the content of international guidelines for cholesterol

lowering and clinical practice in China. With respect to the treatment intensity of

statins, it is defined as high-intensity statin therapy (intensive cholesterol lowering

treatment) when the LDL-C value decreased by more than 50% compared with the

baseline LDL-C value after treatment, and moderate-intensity statin therapy when

there is 30% to 50% of decrease [612, 617].

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 164

Recommendations

It is suggested that LDL-C < 1.8mmol/L (70mg/dL) should be used as a reference

target for cholesterol lowering therapy (Class IIa, Level of Evidence C).

(III) Patients who can significantly benefit from intensive cholesterol-lowering

treatment

In the population with clinical atherosclerotic cardiovascular disease (ASCVD),

patients with LDL-C of 190 mg/dl and above before treatment, 40-75 years old

diabetes patients with LDL-C levels of 70-189 mg/dl without ASCVD, non-diabetes

patients without ASCVD, but with a 10-year ASCVD risk ≥ 7.5% and LDL-C levels

between 70 and 189 mg/dl are those who can benefit from cholesterol-lowering

treatment the most [612].

As a study on non-cardiac ischemic cerebrovascular disease or TIA, patients with

different etiological subtypes, ages, gender, baseline cholesterol levels, with or

without carotid atherosclerosis and diabetes, and diabetes in subgroups of SPARCL

study, all can benefit from intensive cholesterol lowering treatment [611], especially for

subgroups with carotid atherosclerosis [618]. A subgroup analysis of stenting and

intensive medical treatment in the prevention of recurrent stroke in patients with

intracranial atherosclerosis (SAMMPRIS study) has indicated that intensive statin

therapy should be used in stroke patients with intracranial atherosclerosis to prevent

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 165

stroke recurrence [619].

For ischemic stroke outpatients with atrial fibrillation, cholesterol-lowering treatment

with statins can also reduce the recurrence rate [620].

High-intensity statin therapy or the combination treatment of statins with PSCK9

inhibitors can reduce the primary endpoint event rate (major vascular events, etc.) by

19% as compared with conventional-intensity statin therapy [621].

Recommendations

[1] Atrial fibrillation is not a reason for not using statins in patients with

ischemic stroke (Class IIa, Level of Evidence B).

[2] High-intensity statins should be started or continued as a first-line treatment

in female and ≤ 75-year-old male with ASCVD, unless there are

contraindications (Class I, Level of Evidence A).

(IV) Cholesterol-lowering treatments other than statins

Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a new target for lipid-

lowering therapy in recent years. PCSK9 monoclonal antibody (mAb) inhibitors can

bind to the close region of the interaction site with LDLR in PCSK9, thus preventing

PCSK9 from interacting with LDLR so that LDL-C can be cleared more by LDLR.

Previous studies have demonstrated at the molecular level that inhibition of PCSK9

expression can effectively reduce LDL-C, and this effect is potent and profound. In

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 166

the FOURIER study (PCSK9 inhibitors in high-risk populations for further

cardiovascular prognosis), 27,564 patients with atherosclerotic cerebrovascular

disease, fasting LDL-C or non-HDL cholesterol elevation who received optimized

lipid-lowering therapy (at least 20 mg of atorvastatin or other statins of equal

strength) were randomly divided into subcutaneous evolocumab group or placebo

group, with median follow-up time of 2.2 years. Evolocumab treatment significantly

reduced composite primary endpoint events (cardiovascular death, myocardial

infarction, stroke, hospitalization due to unstable angina pectoris, hospitalization for

coronary recanalization) (9.8% vs. placebo group 11.3%; HR=0.85, 95% CI: 0.79-

0.92); and could reduce composite secondary endpoint events (cardiovascular death,

myocardial infarction or stroke) (5.9% vs. placebo group 7.4%; HR=0.80, 95% CI:

0.73-0.88) [622].

A meta-analysis including 23 randomized controlled studies has indicated that lipid-

lowering treatment with niacin can’t reduce cardiovascular death, disability, and non-

disabling stroke [623]. A multi-centre, randomized, controlled, single-blind clinical trial

conducted by domestic scholars has explored the treatment of AIS by reducing

cholesterol levels via biofilm in combination with delipid extracorporeal lipoprotein

filter (DELP), which has indicated that it can significantly improve neurological

function and 90-day prognosis, especially in stroke patients with high blood lipid

level, with no obvious adverse reactions [624].

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 167

Recommendations

In atherosclerotic ischemic stroke patients who have already optimized statins,

measuring blood cholesterol levels may help identify those who can benefit from

PCSK9 treatment (Class IIb, Level of Evidence B).

(V) Combined lipid-lowering treatment

There is no sufficient evidence for the effectiveness of monotherapy of ezetimibe or

ezetimibe in combination with statins in the lipid-lowering treatment compared with

statins. The transaminase test should be perfected before the application of ezetimibe,

and the transaminase level should be monitored during application. If ALT continues

to exceed the normal upper limit by 3 times, the application of ezetimibe should be

discontinued. In the Study of Heart and Renal Protection (SHARP), 10mg of

Ezetimibe in combination with 20mg simvastatin could reduce LDL-C levels by 23%

in patients who received dialysis treatment and 33% in patients who didn’t receive

dialysis treatment compared with placebo [625]. In the study of the Simvastatin and

Ezetimibe in Aortic Stenosis (SEAS), the incidence of composite endpoint events

including ischemic stroke was not reduced in simvastatin + ezetimibe group compared

with that in placebo group [626]. In the IM Proved Reduction of Outcomes: Vytorin

Efficacy International Trial (IMPROVE-IT), the incidence of composite events

including ischemic stroke in patients with acute coronary syndrome in

ezetimibe/simvastatin group was effectively reduced compared with that of placebo

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 168

/simvastatin group in the median 6-year long-term follow-up (the incidence of

ischemic stroke decreased by 21%, P=0.008), which indicated that ezetimibe can

effectively improve the prognosis of patients with acute coronary syndrome [627].

Recommendations

For patients with poor lipid-lowering effect or intolerable of statins, lipid

lowering treatment can be combined with ezetimibe but regular monitoring of

transaminase and physical examination should be done (Class IIb, Level of

Evidence B).

See Supplemental Table 15 for the comparison table of doses of lipid-lowering drugs.

(VI) Instructors of statin cholesterol lowering treatment with statins and patient

compliance

A study included secondary prevention of non-cardiac embolism ischemic stroke has

indicated that although there is no difference in the overall incident rate 12-24 months

after ischemic attack, the guidance group of specialists and nurses can better reduce

lipid level, which is reflected in the reduction degree of LDL-C [628]. A meta-analysis

based on observational studies has shown that the application of statins in hospitals is

associated with good functional prognosis [629]. A retrospective study assessing

compliance of patients after initiating statins for the treatment of ischemic stroke for 3

months indicated that these included patients maintained high medication compliance

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 169

and achieved somewhat good functional prognosis 3 months after discharge [630]. In a

large cohort study with analysis of 12 years of studies, it was found that increased

stroke recurrence would appear within 1 year in stroke patients who were prescribed

lipid-lowering statins at the time of discharge if they stopped taking statins within 2-6

months after a stroke event [631].

Recommendations

ASCVD patients with ischemic stroke and other complications should be

managed through lifestyle improvements, dietary advice, and drug treatment

(Class I, Level of Evidence A).

(VII) Safety of application of high-intensity statins

High-intensity statins should be used with caution in the elderly, patients with

impaired liver function, renal insufficiency or high-risk populations with adverse

reactions that could potentially interact with combined drugs [632]. In consideration of

the safety of statin therapy, comprehensive assessment on the types and doses of

statins should be conducted in male, non-pregnant and non-lactating female based on

patients’ characteristics, risk of coronary atherosclerotic heart disease (calculated

using data from multiple cohorts), and potential adverse reactions. For patients with

multiple comorbidities, including renal insufficiency, liver insufficiency, previous

statin intolerance or muscle disease, unexplainable elevation of transaminase

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 170

more than three times the normal value, statin metabolism affected by the patient’s

own cause or drug combination, and patients over 75 years old, medium-intensity

statins are preferred to high-intensity statins. Other factors influencing the selection of

statin intensity include, but are not limited to, history of haemorrhagic stroke and the

Asian population [612, 633-634]. In the above population, the application of statins in

combination with ezetimibe is safe, which can achieve better lipid-lowering effects

and effectively improve cardiovascular prognosis, especially in patients with a history

of ischemic stroke [635-638].

Recommendations

[1] For ASCVD patients, it was originally intended to be treated with high-

intensity statins. But when patients have contraindications or possible adverse

reactions to statins, moderate intensity statins should be used as a second option.

(Class I, Level of Evidence A).

[2] For clinical ASCVD patients over 75 years old, the benefits of reducing

ASCVD risk, adverse drug reactions, drug-drug interactions and patient’s

wishes should be evaluated when initiating moderate or high-intensity statins. It

is reasonable to continue statins in patients who can tolerate (Class IIb, Level of

Evidence C).

See Figure 10 for the management process of lipid-lowering in patients with acute

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 171

ischemic stroke.

III. Management of abnormal glucose metabolism

Hyperglycaemia is common in patients with AIS. Multiple studies have shown that

more than 40% of AIS patients show elevated blood glucose on admission, and most

have a previous history of diabetes [639- 640]. Blood glucose elevation in stroke patients

may be related to the non-fasting state, as well as impaired glucose metabolism under

stress. Multiple observational studies have shown that blood glucose elevation at

admission and in hospital is associated with poor clinical prognosis [641-642]. In patients

receiving intravenous thrombolytic therapy, hyperglycaemia may be associated with

symptomatic bleeding conversion and poor prognosis [100, 643-644]. In addition, multiple

studies have indicated that AIS is associated with poor prognosis caused by massive

cerebral infarction [645-647]. Although many observational studies have shown that

elevated AIS blood glucose is associated with poor prognosis, it is uncertain whether

there is a direct causal connection between the two.

It is generally accepted that hyperglycaemia should be controlled after stroke, but

there are only a few randomized controlled trials on hypoglycaemic measures and

target blood glucose values [499, 648-651], and no final conclusion has been reached. It is

reasonable to control blood glucose at 140-180 mg/dl based on previous guidelines.

The incidence of hypoglycaemia is low after stroke. Although there is no clinical trial

on hyperglycaemic treatment, hypoglycaemia should be corrected as soon as possible

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 172

because it can directly cause cerebral ischemic injury and aggravated oedema,

resulting in poor prognosis.

There are several large-scale RCT studies on intensified blood glucose management

to reduce the risk of stroke and cardiovascular disease.

The ACCORD study [652] has evaluated whether intensive control of glycated

haemoglobin to a normal target value can reduce cardiovascular disease events in type

2 diabetes patients with cardiovascular disease or cardiovascular risk factors. In this

study, a total of 10,251 patients with average HbA1c of 8.1% were randomly divided

into intensive treatment group using multiple drugs including insulin and oral

hypoglycaemic drugs (target HbA1c value < 6.0%) or standard treatment group

(target value: 7.0%-7.9%). As the death rate due to any cause was somewhat high in

the intensive treatment group, it was stopped early (HR=1.22, 95% CI: 1.01-1.46,

P=0.04). Although the average HbA1c value decreased from baseline 8.1% to 6.7%

(intensive treatment group) and 7.5% (control group) at 4 months, the primary

endpoint risk associated with intensive hypoglycaemic treatment (non-fatal MI, non-

fatal stroke or cardiovascular death) didn’t decrease (6.9% vs. 7.2%, HR = 0.90, 95%

CI: 0.78-1.04, P = 0.16). Patients in the intensive treatment group required more

frequent glucose-lowering drugs as adjuvant therapy (10.5% vs. 3.5%).

Another study evaluated the role of intensive glucose control in diabetic patients with

poor control. After five to six years of follow-up, HbA1c values of patients in the

intensive glycemic control group were significantly reduced, and no significant

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 173

differences in the primary and secondary endpoints were found between the two

groups, including stroke risk (the number of events, 26 vs. 36, HR=0.78, CI: 0.48-

1.28) or TIA (19 vs.13, HR=1.48, 95% CI: 0.73-2.99). The incidence of

hypoglycaemic event increased significantly in the intensive treatment group [653].

In the ADVANCE study, 11,140 patients with type 2 diabetes were randomly divided

into standard glucose-lowering group and intensive glucose-lowering group. The

HbA1c value of the intensive glucose-lowering group was reduced to at least less than

6.5% by drugs. After 5 years of follow-up, it was found that the average HbA1c value

of the intensive treatment group was lower than that of the standard group (6.5% vs.

7.3%) [654]. The incidence of major complications such as macrovascular and

microvascular events in intensive treatment group was reduced (18.1% vs. 20.0%,

HR=0.90, 95%CI: 0.77-0.97, P=0.01). There was no significant difference in

mortality risk between the two groups (HR=0.93, 95% CI 0.83-1.06, P=0.28).

A meta-analysis covering the results of the above three studies evaluated the role of

intensive glucose-lowering treatment in the prevention of vascular events in patients

with type 2 diabetes, with an average follow-up of 5 years. The average HbA1c values

were 6.6% (intensive group) and 7.4% (control group [655]. The incidences of all-cause

death risk, stroke and cardiovascular mortality were not reduced in the intensive

glucose-lowering treatment; however, the incidence of non-fatal myocardial infarction

was significantly reduced by 14% (RR=0.86, 95% CI: 0.77-0.97, P=0.015).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 174

Recommendations

[1] The prognosis of persistent hyperglycaemia in AIS patients within 24 hours

after onset is worse than that of normal blood glucose. So, it is reasonable to

control blood glucose to the range between 140~180mg/dL. At the same time,

blood sugar should be closely monitored to prevent hypoglycaemia (Class IIa,

Level of Evidence C).

[2] Treatment should be given to AIS patients with hypoglycaemia (blood glucose

< 60mg/dL) (Class I, Level of Evidence C).

[3] It is recommended that ischemic stroke or TIA patients with diabetes should

be evaluated and given the best management guideline (Class I, Level of

Evidence A).

[4] For all inpatients/outpatients with ischemic stroke or TIA, rapid blood

glucose, 2 hours postprandial blood glucose, glycosylated haemoglobin or 75g

oral glucose tolerance test are recommended for screening diabetes mellitus

(Class IIa, Level of Evidence C).

[5] Lifestyle and/or drug intervention in patients with diabetes or prediabetes

can reduce ischemic stroke or TIA events. The recommended treatment target

for glycosylated haemoglobin is ≤ 7% (Class I, Level of Evidence B).

[6] The hypoglycaemic regimen should consider the clinical characteristics of

patients and the safety of drugs. To set up individual blood glucose control

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 175

targets, one should be vigilant against the harm caused by hypoglycaemic events

(Class IIa, Level of Evidence B).

IV. Management of other risk factors

Many studies have shown that smoking is an independent risk factor for ischemic

stroke [656-660].A multicentre prospective study on Chinese intracranial atherosclerosis

(CICAS) shows that smoking is dose-related to the occurrence of extracranial

atherosclerosis, but not related to intracranial atherosclerosis [661].

A large number of studies have found that Obstructive Sleep Apnoea is associated

with stroke. Obstructive Sleep Apnoea is common in stroke patients and is associated

with a high incidence of the following events, including cardiovascular and

cerebrovascular events, poor prognosis and higher mortality. Continuous positive

pressure ventilation is still the most effective method to treat apnoea. However, a RCT

study shows that continuous positive pressure ventilation for patients with moderate

to severe apnoea has no benefit in preventing cardiovascular events or deaths in

patients with a history of stroke [662]. Therefore, routine screening of apnoea for

secondary prevention of cardiovascular events or death is not helpful for all AIS

patients.

The National Institute of Abuse and Alcoholism defines heavy drinking for

men as drinking more than 4 units per day or more than 14 units per week, and

defines heavy drinking for women as drinking more than 3 units per day or more than

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 176

7 units per week. So far, the correlation between alcohol intake and stroke is still

controversial. Many research results show that heavy drinking is a risk factor for

various types of stroke [663-669].

In general, a large number of observational studies show that light to moderate

drinking is associated with overall and ischemic stroke risk reduction, while heavy

drinking increases the stroke risk. Many prospective randomized clinical trials show

that heavy drinking can increase the risk of stroke but a small amount of drinking will

reduce the risk of stroke, which lacks ethical support, because alcohol dependence is a

major health problem.

Three large sample retrospective studies have found that hormone replacement

therapy have no significant correlation with the occurrence, severity and prognosis of

stroke [670-672]. Cheetham[673] found in a retrospective cohort study with a sample size

of 8808 that men receiving androgen replacement therapy had a lower incidence of

cardiovascular events than men who had never received such therapy. Sidney found in

a retrospective cohort study that the incidence of thromboembolism events in oral

contraceptives was higher than that in non-oral contraceptives [674].

In recent years, the research on the relationship between oral contraceptives/hormone

replacement therapy and stroke has shown a significant growth trend in China.

Specifically, four retrospective cohort studies have [675-678] found that oral

contraceptives are related to the increased risk of cerebral haemorrhage, especially

when patients have a history of hypertension.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 177

Some studies have proved that there is a causal relationship between acute narcotic

drug use and the risk of early ischemic stroke, rather than previous use of narcotic

drugs [679]. In addition, studies have shown that people with stroke related to narcotic

drug abuse are younger, tend to have a smoking history, and have less traditional risk

factors such as hypertension, diabetes, and hyperlipidaemia [680-682].

Two large-sample meta-analyses based on population cohort studies found that

reducing homocysteine by 25% can reduce stroke risk by 11%-16% [683-684].However,

clinical trials of folic acid supplementation for secondary prevention of CVD or

stroke have not found that supplementation of homocysteine-reducing vitamins can

reduce the risk of recurrent stroke. Currently, there is a lack of large-sample studies of

homocysteine-related genes (MTHFR 677C- > T) and stroke risk [685].The Vitamin

Intervention for Stroke Prevention (VISP) study randomly divided the non-cardiac

stroke patients into groups, and the patients with mild to moderate

hyperhomocysteinemia received high-dose or low-dose vitamin therapy for 2 years.

The results showed that the risk of stroke was related to the homocysteine level, the

average reduction of the homocysteine level in the high dose vitamin treatment group

was larger, but the risk of stroke did not decrease [686]. The "Vitamins to Prevent

Stroke" trial (VITATOPS) also failed to prove that vitamin therapy can prevent stroke,

myocardial infarction or vascular death in patients with recent stroke or TIA [687].

Recommendations

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 178

[1] Healthcare staff should strongly recommend that all AIS patients who have

smoked in the past year quit smoking (Class I, Level of Evidence C).

[2] For AIS patients who smokes, consider to starting intervention measures

combined with drug therapy and behavioural support during hospitalization

(Class IIb, Level of Evidence B).

[3] Routine screening for obstructive sleep apnoea in patients with recent

ischemic stroke is not recommended (Class III, Level of Evidence B).

[4] For alcohol drinkers, it may be reasonable for men to drink ≤ 2 units and

non-pregnant women to drink ≤ 1 unit per day (Class IIb, Level of Evidence B).

[5] The relationship between oral contraceptives and stroke needs to be further

confirmed in prospective studies. Oral contraceptives may be associated with

haemorrhagic stroke, which is more pronounced in patients with hypertension.

So oral contraceptive is not recommended for patients with hypertension (Class

III, Level of Evidence C).

[6] The relationship between drug use and stroke needs to be further studied.

Acute drug use may be a risk factor for stroke and a factor for poor prognosis

(Class III, Level of Evidence C).

[7] For patients with recent ischemic stroke or TIA and mild to moderate

increase in blood homocysteine, Folic acid, vitamin B6 and vitamin B12

supplementation can reduce the level of homocysteine. There is not enough

evidence to support the practice of reducing homocysteine levels to reduce the

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 179

risk of stroke recurrence (Class IIb, Level of Evidence B).

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 180

Supplemental Reference

[1] RUBIN M N, BARRETT K M. What to do With Wake-Up Stroke.

Neurohospitalist, 2015, 5(3): 161-172.

[2] THOMALLA G, BOUTITIE F, FIEBACH J B, et al. Stroke With Unknown Time

of Symptom Onset: Baseline Clinical and Magnetic Resonance Imaging Data of the

First Thousand Patients in WAKE-UP (Efficacy and Safety of MRI-Based

Thrombolysis in Wake-Up Stroke: A Randomized, Doubleblind, Placebo-Controlled

Trial). Stroke, 2017, 48(3): 770-773.

[3] THOMALLA G, SIMONSEN C Z, BOUTITIE F, et al. MRI-Guided

Thrombolysis for Stroke with Unknown Time of Onset. New England Journal of

Medicine, 2018,

[4] ROST N S, MASRUR S, PERVEZ M A, et al. Unsuspected coagulopathy rarely

prevents IV thrombolysis in acute ischemic stroke. Neurology, 2009, 73(23): 1957-

1962.

[5] CUCCHIARA B L, JACKSON B, WEINER M, et al. Usefulness of checking

platelet count before thrombolysis in acute ischemic stroke. Stroke, 2007, 38(5):

1639-1640.

[6] OHSHITA T, IMAMURA E, NOMURA E, et al. Hypoglycaemia with focal

neurological signs as stroke mimic: Clinical and neuroradiological characteristics. J

Neurol Sci, 2015, 353(1-2): 98-101.

[7] AGHAEBRAHIM A, STREIB C, RANGARAJU S, et al. Streamlining door to

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 181

recanalization processes in endovascular stroke therapy. 2017, 9(4): 340-345.

[8] MESSE S R, KHATRI P, REEVES M J, et al. Why are acute ischemic stroke

patients not receiving IV tPA? Results from a national registry. Neurology, 2016,

87(15): 1565-1574.

[9] ZAIDI S F, SHAWVER J, ESPINOSA MORALES A, et al. Stroke care: initial

data from a county-based bypass protocol for patients with acute stroke. J Neurointerv

Surg, 2017, 9(7): 631-635.

[10] LEES K R, EMBERSON J, BLACKWELL L, et al. Effects of Alteplase for

Acute Stroke on the Distribution of Functional Outcomes: A Pooled Analysis of 9

Trials. Stroke; a journal of cerebral circulation, 2016, 47(9): 2373-2379.

[11] WANG Y, LIAO X, ZHAO X, et al. Using recombinant tissue plasminogen

activator to treat acute ischemic stroke in China: analysis of the results from the

Chinese National Stroke Registry (CNSR). Stroke; a journal of cerebral circulation,

2011, 42(6): 1658-1664.

[12] LO E H. A new penumbra: transitioning from injury into repair after stroke. Nat

Med, 2008, 14(5): 497-500.

[13] Tissue for Acute Ischemic Stroke. The New England

journal of medicine, 1995, 333(24): 1581-1588.

[14] HACKE W, KASTE M, FIESCHI C, et al. Randomised double-blind placebo-

controlled trial of thrombolytic therapy with intravenous alteplase in acute ischaemic

stroke (ECASS II). The Lancet, 1998, 352(9136): 1245-1251.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 182

[15] MARKS M P, HEIT J J, LANSBERG M G, et al. Endovascular Treatment in the

DEFUSE 3 Study. Stroke, 2018, 49(8): 2000-2003.

[16] LUO S, YANG L, WANG L. Comparison of susceptibility-weighted and

perfusion-weighted magnetic resonance imaging in the detection of penumbra in

acute ischemic stroke. J Neuroradiol, 2015, 42(5): 255-260.

[17] NIIBO T, OHTA H, YONENAGA K, et al. Arterial spin-labeled perfusion

imaging to predict mismatch in acute ischemic stroke. Stroke, 2013, 44(9): 2601-

2603.

[18] HARSTON G W J, TEE Y K, BLOCKLEY N, et al. Identifying the ischaemic

penumbra using pH-weighted magnetic resonance imaging. Brain : a journal of

neurology, 2015, 138(Pt 1): 36-42.

[19] AN H, FORD A L, CHEN Y, et al. Defining the ischemic penumbra using

magnetic resonance oxygen metabolic index. Stroke, 2015, 46(4): 982-988.

[20] LANSBERG M G, THIJS V N, HAMILTON S, et al. Evaluation of the clinical-

diffusion and perfusion-diffusion mismatch models in DEFUSE. Stroke, 2007, 38(6):

1826-1830.

[21] LANSBERG M G, CEREDA C W, MLYNASH M, et al. Response to

endovascular reperfusion is not time-dependent in patients with salvageable tissue.

Neurology, 2015, 85(8): 708-714.

[22] OGATA T, NAGAKANE Y, CHRISTENSEN S, et al. A topographic study of the

evolution of the MR DWI/PWI mismatch pattern and its clinical impact: a study by

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 183

the EPITHET and DEFUSE Investigators. Stroke, 2011, 42(6): 1596-1601.

[23] TSAI J P, MLYNASH M, CHRISTENSEN S, et al. Time From Imaging to

Endovascular Reperfusion Predicts Outcome in Acute Stroke. Stroke, 2018, 49(4):

952-957.

[24] MENJOT DE CHAMPFLEUR N, SAVER J L, GOYAL M, et al. Efficacy of

Stent-Retriever Thrombectomy in Magnetic Resonance Imaging Versus Computed

Tomographic Perfusion-Selected Patients in SWIFT PRIME Trial (Solitaire FR With

the Intention for Thrombectomy as Primary Endovascular Treatment for Acute

Ischemic Stroke). Stroke, 2017, 48(6): 1560-1566.

[25] CAMPBELL B C V, MITCHELL P J, KLEINIG T J, et al. Endovascular Therapy

for Ischemic Stroke with Perfusion-Imaging Selection. The New England journal of

medicine, 2015, 372(11): 1009-1018.

[26] NOGUEIRA R G, JADHAV A P, HAUSSEN D C, et al. Thrombectomy 6 to 24

Hours after Stroke with a Mismatch between Deficit and Infarct. The New England

journal of medicine, 2018, 378(1): 11-21.

[27] THOMALLA G, FIEBACH J B, ØSTERGAARD L, et al. A multicenter,

randomized, double-blind, placebo-controlled trial to test efficacy and safety of

magnetic resonance imaging-based thrombolysis in wake-up stroke (WAKE-UP).

International journal of stroke : official journal of the International Stroke Society,

2014, 9(6): 829-836.

[28] THOMALLA G, SIMONSEN C Z, BOUTITIE F, et al. MRI-Guided

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 184

Thrombolysis for Stroke with Unknown Time of Onset. The New England journal of

medicine, 2018, 379(7): 611-622.

[29] KIDWELL C S, WINTERMARK M, DE SILVA D A, et al. Multiparametric MRI

and CT models of infarct core and favorable penumbral imaging patterns in acute

ischemic stroke. Stroke, 2013, 44(1): 73-79.

[30] KIDWELL C S, JAHAN R, GORNBEIN J, et al. A trial of imaging selection and

endovascular treatment for ischemic stroke. The New England journal of medicine,

2013, 368(10): 914-923.

[31] HUANG X, CHERIPELLI B K, LLOYD S M, et al. Alteplase versus

tenecteplase for thrombolysis after ischaemic stroke (ATTEST): a phase 2,

randomised, open-label, blinded endpoint study. The Lancet Neurology, 2015, 14(4):

368-376.

[32] ALBERS G W, VON KUMMER R, TRUELSEN T, et al. Safety and efficacy of

desmoteplase given 3-9 h after ischaemic stroke in patients with occlusion or high-

grade stenosis in major cerebral arteries (DIAS-3): a double-blind, randomised,

placebo-controlled phase 3 trial. The Lancet Neurology, 2015, 14(6): 575-584.

[33] WARDLAW J M, MURRAY V, BERGE E, et al. Thrombolysis for acute

ischaemic stroke. The Cochrane database of systematic reviews, 2014, 7): Cd000213.

[34] PARSONS M, SPRATT N, BIVARD A, et al. A randomized trial of tenecteplase

versus alteplase for acute ischemic stroke. The New England journal of medicine,

2012, 366(12): 1099-1107.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 185

[35] HACKE W, FURLAN A J, AL-RAWI Y, et al. Intravenous desmoteplase in

patients with acute ischaemic stroke selected by MRI perfusion-diffusion weighted

imaging or perfusion CT (DIAS-2): a prospective, randomised, double-blind, placebo-

controlled study. The Lancet Neurology, 2009, 8(2): 141-150.

[36] DAVIS S M, DONNAN G A, PARSONS M W, et al. Effects of alteplase beyond

3 h after stroke in the Echoplanar Imaging Thrombolytic Evaluation Trial

(EPITHET): a placebo-controlled randomised trial. The Lancet Neurology, 2008,

7(4): 299-309.

[37] FURLAN A J, EYDING D, ALBERS G W, et al. Dose Escalation of

Desmoteplase for Acute Ischemic Stroke (DEDAS): evidence of safety and efficacy 3

to 9 hours after stroke onset. Stroke; a journal of cerebral circulation, 2006, 37(5):

1227-1231.

[38] ALBERS G W, THIJS V N, WECHSLER L, et al. Magnetic resonance imaging

profiles predict clinical response to early reperfusion: the diffusion and perfusion

imaging evaluation for understanding stroke evolution (DEFUSE) study. Annals of

neurology, 2006, 60(5): 508-517.

[39] HACKE W, ALBERS G, AL-RAWI Y, et al. The Desmoteplase in Acute

Ischemic Stroke Trial (DIAS): a phase II MRI-based 9-hour window acute stroke

thrombolysis trial with intravenous desmoteplase. Stroke; a journal of cerebral

circulation, 2005, 36(1): 66-73.

[40] BERKHEMER O A, FRANSEN P S, BEUMER D, et al. A randomized trial of

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 186

intraarterial treatment for acute ischemic stroke. The New England journal of

medicine, 2015, 372(1): 11-20.

[41] GOYAL M, DEMCHUK A M, MENON B K, et al. Randomized assessment of

rapid endovascular treatment of ischemic stroke. The New England journal of

medicine, 2015, 372(11): 1019-1030.

[42] SAVER J L, GOYAL M, BONAFE A, et al. Stent-retriever thrombectomy after

intravenous t-PA vs. t-PA alone in stroke. The New England journal of medicine,

2015, 372(24): 2285-2295.

[43] JOVIN T G, CHAMORRO A, COBO E, et al. Thrombectomy within 8 hours

after symptom onset in ischemic stroke. The New England journal of medicine, 2015,

372(24): 2296-2306.

[44] BRACARD S, DUCROCQ X, MAS J L, et al. Mechanical thrombectomy after

intravenous alteplase versus alteplase alone after stroke (THRACE): a randomised

controlled trial. The Lancet Neurology, 2016, 15(11): 1138-1147.

[45] ALBERS G W, MARKS M P, KEMP S, et al. Thrombectomy for Stroke at 6 to

16 Hours with Selection by Perfusion Imaging. 2018, 378(8): 708-718.

[46] HILL M D, DEMCHUK A M, GOYAL M, et al. Alberta Stroke Program early

computed tomography score to select patients for endovascular treatment:

Interventional Management of Stroke (IMS)-III Trial. Stroke; a journal of cerebral

circulation, 2014, 45(2): 444-449.

[47] MACCALLUM C, CHURILOV L, MITCHELL P, et al. Low Alberta Stroke

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 187

Program Early CT score (ASPECTS) associated with malignant middle cerebral

artery infarction. Cerebrovasc Dis, 2014, 38(1): 39-45.

[48] YOO A J, ZAIDAT O O, CHAUDHRY Z A, et al. Impact of pretreatment

noncontrast CT Alberta Stroke Program Early CT Score on clinical outcome after

intra-arterial stroke therapy. Stroke; a journal of cerebral circulation, 2014, 45(3):

746-751.

[49] YEO L L, PALIWAL P, TEOH H L, et al. Assessment of intracranial collaterals

on CT angiography in anterior circulation acute ischemic stroke. AJNR American

journal of neuroradiology, 2015, 36(2): 289-294.

[50] TAN B Y, WAN-YEE K, PALIWAL P, et al. Good Intracranial Collaterals Trump

Poor ASPECTS (Alberta Stroke Program Early CT Score) for Intravenous

Thrombolysis in Anterior Circulation Acute Ischemic Stroke. Stroke; a journal of

cerebral circulation, 2016, 47(9): 2292-2298.

[51] YOO A J, BERKHEMER O A, FRANSEN P S S, et al. Effect of baseline Alberta

Stroke Program Early CT Score on safety and efficacy of intra-arterial treatment: a

subgroup analysis of a randomised phase 3 trial (MR CLEAN). The Lancet

Neurology, 2016, 15(7): 685-694.

[52] LI W, LI S, DAI M, et al. Comparisons of ASPECTS 5 and 6 for endovascular

treatment in anterior circulation occlusive stroke. Interventional neuroradiology :

journal of peritherapeutic neuroradiology, surgical procedures and related

neurosciences, 2017, 23(5): 516-520.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 188

[53] XU C, TAO A, WANG Z, et al. A Retrospective Study of Clinical Outcomes After

Endovascular Treatment in Acute Ischemic Stroke Patients with Complete Anterior

Circulation Infarction in the Absence of Multimodal Computed Tomography. World

neurosurgery, 2017, 108(460-464.

[54] NAYLOR J, CHURILOV L, CHEN Z, et al. Reliability, Reproducibility and

Prognostic Accuracy of the Alberta Stroke Program Early CT Score on CT Perfusion

and Non-Contrast CT in Hyperacute Stroke. Cerebrovasc Dis, 2017, 44(3-4): 195-

202.

[55] SALLUSTIO F, MOTTA C. CT Angiography ASPECTS Predicts Outcome Much

Better Than Noncontrast CT in Patients with Stroke Treated Endovascularly. 2017,

38(8): 1569-1573.

[56] HUI F K, OBUCHOWSKI N A, JOHN S, et al. ASPECTS discrepancies between

CT and MR imaging: analysis and implications for triage protocols in acute ischemic

stroke. Stroke, 2017, 9(3): 240-243.

[57] DESILLES J P, CONSOLI A, REDJEM H, et al. Successful Reperfusion With

Mechanical Thrombectomy Is Associated With Reduced Disability and Mortality in

Patients With Pretreatment Diffusion-Weighted Imaging-Alberta Stroke Program

Early Computed Tomography Score

2017, 48(4): 963-969.

[58] LYDEN P, RAMAN R, LIU L, et al. National Institutes of Health Stroke Scale

certification is reliable across multiple venues. Stroke, 2009, 40(7): 2507-2511.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 189

[59] WANG D L, PENG Y B, XING L, et al. The use of national institute of health

stroke scale and establishment of the regression model in acute cerebral thrombosis.

Journal of North China Coal Medical College, 2007, 03): 297-298.

[60] NATIONAL INSTITUTE OF NEUROLOGICAL D, STROKE RT P A S S G.

Tissue plasminogen activator for acute ischemic stroke. N Engl J Med, 1995, 333(24):

1581-1587.

[61] HACKE W, KASTE M, BLUHMKI E, et al. Thrombolysis with alteplase 3 to 4.5

hours after acute ischemic stroke. N Engl J Med, 2008, 359(13): 1317-1329.

[62] GROUP I S T C, SANDERCOCK P, WARDLAW J M, et al. The benefits and

harms of intravenous thrombolysis with recombinant tissue plasminogen activator

within 6 h of acute ischaemic stroke (the third international stroke trial [IST-3]): a

randomised controlled trial. Lancet, 2012, 379(9834): 2352-2363.

[63] WARDLAW J M, MURRAY V, BERGE E, et al. Recombinant tissue

plasminogen activator for acute ischaemic stroke: an updated systematic review and

meta-analysis. Lancet, 2012, 379(9834): 2364-2372.

[64] BUTCHER K, CHRISTENSEN S, PARSONS M, et al. Postthrombolysis Blood

Pressure Elevation Is Associated With Haemorrhagic Transformation. Stroke, 2010,

41(1): 72-77.

[65] PERINI F, DE BONI A, MARCON M, et al. Systolic blood pressure contributes

to intracerebral haemorrhage after thrombolysis for ischemic stroke. Journal of the

Neurological Sciences, 2010, 297(1-2): 52-54.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 190

[66] TONI D, AHMED N, ANZINI A, et al. Intravenous thrombolysis in young stroke

patients Results from the SITS-ISTR. Neurology, 2012, 78(12): 880-887.

[67] MAZYA M, EGIDO J A, FORD G A, et al. Predicting the Risk of Symptomatic

Intracerebral Haemorrhage in Ischemic Stroke Treated With Intravenous Alteplase

Safe Implementation of Treatments in Stroke (SITS) Symptomatic Intracerebral

Haemorrhage Risk Score. Stroke, 2012, 43(6): 1524-1531.

[68] WU W, HUO X C, ZHAO X Q, et al. Relationship between Blood Pressure and

Outcomes in Acute Ischemic Stroke Patients Administered Lytic Medication in the

TIMS-China Study. Plos One, 2016, 11(2):

[69] ENDO K, KARIO K, KOGA M, et al. Impact of Early Blood Pressure Variability

on Stroke Outcomes After Thrombolysis The SAMURAI rt-PA Registry. Stroke,

2013, 44(3): 816-+.

[70] WALTIMO T, HAAPANIEMI E, SURAKKA I L, et al. Post-thrombolytic blood

pressure and symptomatic intracerebral haemorrhage. European Journal of Neurology,

2016, 23(12): 1757-1762.

[71] LIU K Q, YAN S Q, ZHANG S, et al. Systolic Blood Pressure Variability is

Associated with Severe Haemorrhagic Transformation in the Early Stage After

Thrombolysis. Translational Stroke Research, 2016, 7(3): 186-191.

[72] AY H, FURIE K L, SINGHAL A, et al. An evidence-based causative

classification system for acute ischemic stroke. Annals of neurology, 2005, 58(5):

688-697.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 191

[73] COUTTS S B, ELIASZIW M, HILL M D, et al. An improved scoring system for

identifying patients at high early risk of stroke and functional impairment after an

acute transient ischemic attack or minor stroke. International journal of stroke :

official journal of the International Stroke Society, 2008, 3(1): 3-10.

[74] WANG X, ROBINSON T G, LEE T H, et al. Low-Dose vs Standard-Dose

Alteplase for Patients With Acute Ischemic Stroke: Secondary Analysis of the

ENCHANTED Randomized Clinical Trial. JAMA neurology, 2017, 74(11): 1328-

1335.

[75] HAN S W, KIM S H, LEE J Y, et al. A new subtype classification of ischemic

stroke based on treatment and etiologic mechanism. European neurology, 2007, 57(2):

96-102.

[76] ILZECKA J, STELMASIAK Z. [Practical significance of ischemic stroke OCSP

(Oxfordshire Community Stroke Project) classification]. Neurol Neurochir Pol, 2000,

34(1): 11-22.

[77] TSIVGOULIS G, ZAND R, KATSANOS A H, et al. Risk of Symptomatic

Intracerebral Haemorrhage After Intravenous Thrombolysis in Patients With Acute

Ischemic Stroke and High Cerebral Microbleed Burden: A Meta-analysis. JAMA

neurology, 2016, 73(6): 675-683.

[78] GAHN G, KDZIALOWSKI B. Combined thrombolysis with abciximab and rtPA

in patients with middle cerebral artery occlusion. Acta Neurologica Scandinavica,

2010, 121(1): 63-66.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 192

[79] NACU A, KVISTAD C E, NAESS H, et al. NOR-SASS (Norwegian

Sonothrombolysis in Acute Stroke Study). Stroke, 2017, 48(2): 335-341.

[80] ADAMS H P, JR., BENDIXEN B H, KAPPELLE L J, et al. Classification of

subtype of acute ischemic stroke. Definitions for use in a multicenter clinical trial.

TOAST. Trial of Org 10172 in Acute Stroke Treatment. Stroke, 1993, 24(1): 35-41.

[81] OGAWA A, MORI E, MINEMATSU K, et al. Randomized trial of intraarterial

infusion of urokinase within 6 hours of middle cerebral artery stroke: the middle

cerebral artery embolism local fibrinolytic intervention trial (MELT) Japan. Stroke,

2007, 38(10): 2633-2639.

[82] INVESTIGATORS I M S S. Combined intravenous and intra-arterial

recanalization for acute ischemic stroke: the Interventional Management of Stroke

Study. Stroke, 2004, 35(4): 904-911.

[83] INVESTIGATORS I I T. The Interventional Management of Stroke (IMS) II

Study. Stroke, 2007, 38(7): 2127-2135.

[84] LEWANDOWSKI C A, FRANKEL M, TOMSICK T A, et al. Combined

intravenous and intra-arterial r-TPA versus intra-arterial therapy of acute ischemic

stroke: Emergency Management of Stroke (EMS) Bridging Trial. Stroke, 1999,

30(12): 2598-2605.

[85] COMPTER A, VAN DER HOEVEN E J, VAN DER WORP H B, et al. Vertebral

artery stenosis in the Basilar Artery International Cooperation Study (BASICS):

prevalence and outcome. Journal of neurology, 2015, 262(2): 410-417.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 193

[86] GOYAL M, MENON B K, VAN ZWAM W H, et al. Endovascular thrombectomy

after large-vessel ischaemic stroke: a meta-analysis of individual patient data from

five randomised trials. Lancet, 2016, 387(10029): 1723-1731.

[87] COUTINHO J, LIEBESKIND D, SLATER L, et al. Combined Intravenous

Thrombolysis and Thrombectomy vs Thrombectomy Alone for Acute Ischemic

Stroke: A Pooled Analysis of the SWIFT and STAR Studies. JAMA neurology, 2017,

74(3): 268-274.

[88] REBELLO L C, HAUSSEN D C, GROSSBERG J A, et al. Early Endovascular

Treatment in Intravenous Tissue Plasminogen Activator-Ineligible Patients. Stroke; a

journal of cerebral circulation, 2016, 47(4): 1131-1134.

[89] LODI Y, REDDY V, PETRO G, et al. Primary acute stroke thrombectomy within

3 h for large artery occlusion (PAST3-LAO): a pilot study. J Neurointerv Surg, 2017,

9(4): 352-356.

[90] GUEDIN P, LARCHER A, DECROIX J P, et al. Prior IV Thrombolysis

Facilitates Mechanical Thrombectomy in Acute Ischemic Stroke. Journal of stroke

and cerebrovascular diseases : the official journal of National Stroke Association,

2015, 24(5): 952-957.

[91] MISTRY E A, MISTRY A M, NAKAWAH M O, et al. Mechanical

Thrombectomy Outcomes With and Without Intravenous Thrombolysis in Stroke

Patients: A Meta-Analysis. Stroke, 2017, 48(9): 2450-2456.

[92] BEHME D, KABBASCH C, KOWOLL A, et al. Intravenous Thrombolysis

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 194

Facilitates Successful Recanalization with Stent-Retriever Mechanical Thrombectomy

in Middle Cerebral Artery Occlusions. Journal of stroke and cerebrovascular

diseases : the official journal of National Stroke Association, 2016, 25(4): 954-959.

[93] ANGERMAIER A, MICHEL P, KHAW A V, et al. Intravenous Thrombolysis and

Passes of Thrombectomy as Predictors for Endovascular Revascularization in

Ischemic Stroke. Journal of stroke and cerebrovascular diseases : the official journal

of National Stroke Association, 2016, 25(10): 2488-2495.

[94] CAMPBELL B C V, MITCHELL P J, CHURILOV L, et al. Tenecteplase versus

Alteplase before Thrombectomy for Ischemic Stroke. N Engl J Med, 2018, 378(17):

1573-1582.

[95] FERRELL A S, BRITZ G W. Developments on the horizon in the treatment of

neurovascular problems. Surg Neurol Int, 2013, 4(Suppl 1): S31-37.

[96] ZAIDAT O O, LAZZARO M A, GUPTA R, et al. Interventional Management of

Stroke III trial: establishing the foundation. J Neurointerv Surg, 2012, 4(4): 235-237.

[97] KIDWELL C S, JAHAN R, GORNBEIN J, et al. A trial of imaging selection and

endovascular treatment for ischemic stroke. N Engl J Med, 2013, 368(10): 914-923.

[98] CICCONE A, VALVASSORI L, NICHELATTI M, et al. Endovascular treatment

for acute ischemic stroke. N Engl J Med, 2013, 368(10): 904-913.

[99] CAST: randomised placebo-controlled trial of early aspirin use in 20,000 patients

with acute ischaemic stroke. CAST (Chinese Acute Stroke Trial) Collaborative

Group. Lancet, 1997, 349(9066): 1641-1649.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 195

[100] CUCCHIARA B, TANNE D, LEVINE S R, et al. A risk score to predict

intracranial haemorrhage after recombinant tissue plasminogen activator for acute

ischemic stroke. Journal of stroke and cerebrovascular diseases : the official journal of

National Stroke Association, 2008, 17(6): 331-333.

[101] SANDERCOCK P A, COUNSELL C, TSENG M C, et al. Oral antiplatelet

therapy for acute ischaemic stroke. Cochrane Database Syst Rev, 2014, 3):

CD000029.

[102] BAIGENT C, BLACKWELL L, COLLINS R, et al. Aspirin in the primary and

secondary prevention of vascular disease: collaborative meta-analysis of individual

participant data from randomised trials. Lancet, 2009, 373(9678): 1849-1860.

[103] FIELDS W, LEMAK N, FRANKOWSKI R, et al. Controlled trial of aspirin in

cerebral ischemia. Circulation, 1980, 62(6 Pt 2): V90-96.

[104] United Kingdom transient ischaemic attack (UK-TIA) aspirin trial: interim

results. UK-TIA Study Group. Br Med J (Clin Res Ed), 1988, 296(6618): 316-320.

[105] GROUP T S C. Swedish Aspirin Low-dose Trial (SALT) of 75 mg aspirin as

secondary prophylaxis after cerebrovascular ischaemic events. Lancet, 1991,

338(8779): 1345–1349.

[106] AUTHORS N. Tissue plasminogen activator for acute ischemic stroke. The

National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group.

New England Journal of Medicine, 1995, 333(24): 1581.

[107] AMARO S, LLULL L, URRA X, et al. Risks and Benefits of Early

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 196

Antithrombotic Therapy after Thrombolytic Treatment in Patients with Acute Stroke.

Plos One, 2013, 8(8): e71132.

[108] DIEDLER J, AHMED N, SYKORA M, et al. Safety of intravenous

thrombolysis for acute ischemic stroke in patients receiving antiplatelet therapy at

stroke onset. Stroke, 2010, 41(2): 288-294.

[109] COMMITTEE C S. A randomised, blinded, trial of clopidogrel versus aspirin in

patients at risk of ischaemic events (CAPRIE). CAPRIE Steering Committee. Lancet,

1996, 348(9038): 1329.

[110] RINGLEB P A, BHATT D L, HIRSCH A T, et al. Benefit of Clopidogrel Over

Aspirin Is Amplified in Patients With a History of Ischemic Events. Stroke, 2004,

35(2): 528.

[111] NIU P P, GUO Z N, HANG J, et al. Antiplatelet regimens in the long-term

secondary prevention of transient ischaemic attack and ischaemic stroke: an updated

network meta-analysis. Bmj Open, 2016, 6(3): e009013.

[112] GURBEL P A, ANTONINO M J, TANTRY U S. Recent developments in

clopidogrel pharmacology and their relation to clinical outcomes. Expert Opinion on

Drug Metabolism & Toxicology, 2009, 5(8): 989-1004.

[113] SURI M F, HUSSEIN H M, ABDELMOULA M M, et al. Safety and

tolerability of 600 mg clopidogrel bolus in patients with acute ischemic stroke:

preliminary experience. Med Sci Monit, 2008, 14(10): Pi39-44.

[114] GURBEL P A, BLIDEN K P, BUTLER K, et al. Randomized double-blind

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 197

assessment of the ONSET and OFFSET of the antiplatelet effects of ticagrelor versus

clopidogrel in patients with stable coronary artery disease: the ONSET/OFFSET

study. Circulation, 2009, 120(25): 2577.

[115] DINICOLANTONIO J J, SEREBRUANY V L. Comparing ticagrelor versus

clopidogrel in patients with a history of cerebrovascular disease: a net clinical harm?

Stroke; a journal of cerebral circulation, 2012, 43(12): 3409.

[116] JOHNSTON S, AMARENCO P, ALBERS G, et al. Ticagrelor versus Aspirin in

Acute Stroke or Transient Ischemic Attack. N Engl J Med, 2016, 375(1): 35-43.

[117] AMARENCO P, ALBERS G W, DENISON H, et al. Efficacy and safety of

ticagrelor versus aspirin in acute stroke or transient ischaemic attack of atherosclerotic

origin: a subgroup analysis of SOCRATES, a randomised, double-blind, controlled

trial. Lancet Neurology, 2017, 16(4): 301-310.

[118] LIU W, LIU R, SUN W, et al. Different impacts of blood pressure variability on

the progression of cerebral microbleeds and white matter lesions. Stroke, 2012,

43(11): 2916-2922.

[119] S. U. Results of the Cilostazol Stroke Prevention Study II (CSPS II): a

randomized controlled trial for the comparison of cilostazol and aspirin in stroke

patients. Rinsho Shinkeigaku, 2010, 50(11): 832-834.

[120] LEE J Y, SUNG K C, CHOI H I. Comparison of aspirin and indobufen in

healthy volunteers. Platelets, 2016, 27(2): 105-109.

[121] LIU J, XU D, XIA N, et al. Anticoagulant Activities of Indobufen, an

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 198

Antiplatelet Drug. Molecules (Basel, Switzerland), 2018, 23(6):

[122] ROGAN J. Indobufen in secondary prevention of transient ischaemic attack.

Multicentre Ischaemic Attack Study Group. J Int Med Res, 1990, 18(3): 240-244.

[123] YANG K, SUN J F. Clinic analysis of treatment of the acute ischemic stroke

with indobufen. China Medicine, 2012, 07(2)

[124] LAVEZZARI M, MILANESI G, SACCHETTI G, et al. Indobufen: Results of a

postmarketing surveillance study on 5,642 cases [M]. 1989.

[125] YANG X, LIU W, CHEN K, et al. Efficacy and safety of indobufen for

prevention and treatment of ischemic cardiovascular and cerebrovascular diseases: a

Meta-analysis. The Chinese Journal of Clinical Pharmacology, 2017, 33(4): 359-362.

[126] INVESTIGATORS A I I S. Abciximab in acute ischemic stroke. A randomized,

double-blind, placebo-controlled, dose-escalation study. Stroke; a journal of cerebral

circulation, 2000, 31(3): 601.

[127] JR A H, MB E, J T, et al. Emergency Administration of Abciximab for

Treatment of Patients With Acute Ischemic Stroke: Results of an International Phase

III Trial Abciximab in Emergency Treatment of Stroke Trial (AbESTT-II). Stroke,

2008, 39(1): 87.

[128] LEE D H, JO K D, KIM H G, et al. Local Intraarterial Urokinase Thrombolysis

of Acute Ischemic Stroke with or without Intravenous Abciximab: A Pilot Study.

Journal of Vascular & Interventional Radiology, 2002, 13(8): 769-773.

[129] KELLERT L, HAMETNER C, ROHDE S, et al. Endovascular Stroke Therapy.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 199

Stroke, 2013, 44(44): e113-e113.

[130] LIN L, LI W, LIU C C, et al. Safety and preliminary efficacy of intravenous

tirofiban in acute ischemic stroke patient without arterial occlusion on neurovascular

imaging studies. Journal of the Neurological Sciences, 2017, 383(175-179.

[131] SIEBLER M, HENNERICI M G, SCHNEIDER D, et al. Safety of Tirofiban in

acute Ischemic Stroke: the SaTIS trial. Stroke; a journal of cerebral circulation, 2011,

42(9): 2388-2392.

[132] DAI Z, LI M, WANG H M, et al. The safety and efficacy of tirofiban on

prevention of vascular reocclusion following mechanical thrombectomy for in situ

thrombosis. Chinese Journal of Neurology, 2017, 6): 440-444.

[133] FENG X W, CHEN Z C, ZHONG G L, et al. Safety of tirofiban in patients with

acute cerebral infarct receiving endovascular therapy. Journal of ZheJiang

University(Medical Science), 2017, 4): 397-404.

[134] LIU Z Q, PENG X Y. Comparison of efficacy between two antiplatelet

therapies in patients with recurrent transient ischemic stroke. China Modern Doctor,

2017, 36): 113-115.

[135] WANG S, LIU M, ZHENG Y Z, et al. Comparison between small dose of

tirofiban and aspirin combined with clopidogrel in the treatment of progressive

stroke in terms of efficacy and safety. Chinese Journal of Practical Nervous

Diseases, 2017, 8): 23-25.

[136] ADEOYE O, SUCHAREW H, KHOURY J, et al. Recombinant tissue-type

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 200

plasminogen activator plus eptifibatide versus recombinant tissue-type plasminogen

activator alone in acute ischemic stroke: propensity score-matched post hoc analysis.

Stroke; a journal of cerebral circulation, 2015, 46(2): 461-464.

[137] ADEOYE O, SUCHAREW H, KHOURY J, et al. Combined Approach to Lysis

Utilizing Eptifibatide and Recombinant Tissue-Type Plasminogen Activator in Acute

Ischemic Stroke-Full Dose Regimen Stroke Trial. Stroke, 2015, 46(9): 2529-2533.

[138] KENNEDY J, HILL M D, RYCKBORST K J, et al. Fast assessment of stroke

and transient ischaemic attack to prevent early recurrence (FASTER): a randomised

controlled pilot trial. The Lancet Neurology, 2007, 6(11): 961-969.

[139] DIENER H C, BOGOUSSLAVSKY J, BRASS L M, et al. Aspirin and

clopidogrel compared with clopidogrel alone after recent ischaemic stroke or transient

ischaemic attack in high-risk patients (MATCH): randomised, double-blind, placebo-

controlled trial. Dkgest of the World Latest Medical Information, 2005, 40(6): 1259-

1259.

[140] INVESTIGATORS T S. Effects of Clopidogrel Added to Aspirin in Patients

with Recent Lacunar Stroke. N Engl J Med, 2012, 367(9): 817-825.

[141] WANG Y, WANG Y, ZHAO X, et al. Clopidogrel with aspirin in acute minor

stroke or transient ischemic attack. N Engl J Med, 2013, 369(1): 11-19.

[142] GE F, LIN H, LIU Y, et al. Dual antiplatelet therapy after stroke or transient

ischaemic attack - how long to treat? The duration of aspirin plus clopidogrel in stroke

or transient ischaemic attack: a systematic review and meta-analysis. European

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 201

Journal of Neurology, 2016, 23(6): 1051-1057.

[143] YI X, LIN J, WANG C, et al. A comparative study of dual versus

monoantiplatelet therapy in patients with acute large-artery atherosclerosis stroke.

Journal of Stroke & Cerebrovascular Diseases the Official Journal of National Stroke

Association, 2014, 23(7): 1975-1981.

[144] JOHNSTON S C, EASTON J D, FARRANT M, et al. Clopidogrel and Aspirin

in Acute Ischemic Stroke and High-Risk TIA. N Engl J Med, 2018,

[145] MILLAN-GUERRERO R, ISAIS-CARDENAS M. [Intravenous dipyridamole

in acute cerebral infarct. Is it efficacious?]. Gaceta medica de Mexico, 1999, 135(4):

391-396.

[146] HAUNGSAITHONG R, UDOMMONGKOL C, NIDHINANDANA S, et al.

The Changes in Mean Platelet Volume after Using of Antiplatelet Drugs in Acute

Ischemic Stroke: A Randomized Controlled Trial. J Med Assoc Thai, 2015, 98(9):

852-857.

[147] CHAIRANGSARIT P, SITHINAMSUWAN P, NIYASOM S, et al. Comparison

between aspirin combined with dipyridamole versus aspirin alone within 48 hours

after ischemic stroke event for prevention of recurrent stroke and improvement of

neurological function: a preliminary study. J Med Assoc Thai, 2005, 88 Suppl 3(S148.

[148] DENGLER R, DIENER H C, SCHWARTZ A, et al. Early treatment with

aspirin plus extended-release dipyridamole for transient ischaemic attack or ischaemic

stroke within 24 h of symptom onset (EARLY trial): a randomised, open-label,

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 202

blinded-endpoint trial. Lancet Neurology, 2010, 9(2): 159.

[149] BATH P M, COTTON D, MARTIN R H, et al. Effect of combined aspirin and

extended-release dipyridamole versus clopidogrel on functional outcome and

recurrence in acute, mild ischemic stroke: PRoFESS subgroup analysis. Stroke, 2010,

41(4): 732.

[150] LIANG T, DENG S R. Observation on efficacy of aspirin and dipyridamole in

prerventing recurrence of ischemic stroke. China Modern Doctor, 2010, 19): 38-39.

[151] BATH P M, WOODHOUSE L J, APPLETON J P, et al. Triple versus guideline

antiplatelet therapy to prevent recurrence after acute ischaemic stroke or transient

ischaemic attack: the TARDIS RCT. Health Technol Assess, 2018, 22(48): 1-76.

[152] BATH P M, WOODHOUSE L J, APPLETON J P, et al. Antiplatelet therapy

with aspirin, clopidogrel, and dipyridamole versus clopidogrel alone or aspirin and

dipyridamole in patients with acute cerebral ischaemia (TARDIS): a randomised,

open-label, phase 3 superiority trial. Lancet, 2018, 391(10123): 850-859.

[153] LU X L, LUO D, YAO X L, et al. dl-3n-Butylphthalide promotes angiogenesis

via the extracellular signal-regulated kinase 1/2 and phosphatidylinositol 3-

kinase/Akt-endothelial synthase signaling pathways. J Cardiovasc

Pharmacol, 2012, 59(4): 352-362.

[154] ZHAO Y, LEE J H, CHEN D, et al. DL-3-n-butylphthalide induced

neuroprotection, regenerative repair, functional recovery and psychological benefits

following traumatic brain injury in mice. Neurochem Int, 2017, 111(82-92.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 203

[155] QIN C, ZHOU P, WANG L, et al. Dl-3-N-butylphthalide attenuates ischemic

reperfusion injury by improving the function of cerebral artery and circulation.

Journal of cerebral blood flow and metabolism : official journal of the International

Society of Cerebral Blood Flow and Metabolism, 2018, 271678X18776833.

[156] CUI L Y, LI S W, ZHANG W W, et al. Effects of dl-3-butylphthalide soft

capsules on treatment of acute ischemlC stroke: multi-center, randomized, double-

blind, double-dummy and aspirin-control study. Chinese Journal of Neurology, 2008,

41(11): 727-730. 8850

[157] Ding D Y, LV C Z, DING M P, et al. A multicenter, randomized,double-blinded

and placebo-controlled study of acute brain infarction treated by humanurinary

kallidinogenase. Chinese Journal of Neurology, 2007, 40(5): 306-310.

[158] WU L R, LIU L, XIONG X Y, et al. Vinpocetine alleviate cerebral

ischemia/reperfusion injury by down-regulating TLR4/MyD88/NF-kappaB signaling.

Oncotarget, 2017, 8(46): 80315-80324.

[159] ZHANG F, YAN C, WEI C, et al. Vinpocetine Inhibits NF-kappaB-Dependent

Inflammation in Acute Ischemic Stroke Patients. Translational stroke research, 2018,

9(2): 174-184.

[160] JEON K I, XU X, AIZAWA T, et al. Vinpocetine inhibits NF-kappaB-dependent

inflammation via an IKK-dependent but PDE-independent mechanism. Proc Natl

Acad Sci U S A, 2010, 107(21): 9795-9800.

[161] ZHENG J, CHEN X. Edaravone offers neuroprotection for acute diabetic stroke

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 204

patients. Irish journal of medical science, 2016, 185(4): 819-824.

[162] YAGI K, KITAZATO K T, UNO M, et al. Edaravone, a free radical scavenger,

inhibits MMP-9-related brain haemorrhage in rats treated with tissue plasminogen

activator. Stroke, 2009, 40(2): 626-631.

[163] YAMAGUCHI T, AWANO H, MATSUDA H, et al. Edaravone with and

without .6 Mg/Kg Alteplase within 4.5 Hours after Ischemic Stroke: A Prospective

Cohort Study (PROTECT4.5). Journal of stroke and cerebrovascular diseases : the

official journal of National Stroke Association, 2017, 26(4): 756-765.

[164] MIYAJI Y, YOSHIMURA S, SAKAI N, et al. Effect of edaravone on favorable

outcome in patients with acute cerebral large vessel occlusion: subanalysis of

RESCUE-Japan Registry. Neurol Med Chir (Tokyo), 2015, 55(3): 241-247.

[165] EHRENREICH H, HASSELBLATT M, DEMBOWSKI C, et al. Erythropoietin

therapy for acute stroke is both safe and beneficial. Molecular medicine (Cambridge,

Mass), 2002, 8(8): 495-505.

[166] ASADI B, ASKARI G R, KHORVASH F, et al. Neuroprotective effects of

erythropoietin in acute ischemic stroke. International journal of preventive medicine,

2013, 4(Suppl 2): S306-312.

[167] EHRENREICH H, WEISSENBORN K, PRANGE H, et al. Recombinant

human erythropoietin in the treatment of acute ischemic stroke. Stroke, 2009, 40(12):

e647-656.

[168] SHU Z M, SHU X D, LI H Q, et al. Ginkgolide B Protects Against Ischemic

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 205

Stroke Via Modulating Microglia Polarization in Mice. CNS Neurosci Ther, 2016,

22(9): 729-739.

[169] DONG Q. Ginkgolide in ischemic stroke patients with large artery athero

sclerosis(GISAA): a randomized D-B, multicenter, placebo-controlled study. British

Heart Diseases, 2019, 2(1): 119-124.

[170] MAZYA M, EGIDO J A, FORD G A, et al. Predicting the risk of symptomatic

intracerebral haemorrhage in ischemic stroke treated with intravenous alteplase: safe

Implementation of Treatments in Stroke (SITS) symptomatic intracerebral

haemorrhage risk score. Stroke, 2012, 43(6): 1524-1531.

[171] GUSEV E I, SKVORTSOVA V I, DAMBINOVA S A, et al. Neuroprotective

effects of glycine for therapy of acute ischaemic stroke. Cerebrovasc Dis, 2000, 10(1):

49-60.

[172] Phase II studies of the glycine antagonist GV150526 in acute stroke : the North

American experience. The North American Glycine Antagonist in Neuroprotection

(GAIN) Investigators. Stroke, 2000, 31(2): 358-365.

[173] LEES K R, ASPLUND K, CAROLEI A, et al. Glycine antagonist (gavestinel)

in neuroprotection (GAIN International) in patients with acute stroke: a randomised

controlled trial. GAIN International Investigators. Lancet, 2000, 355(9219): 1949-

1954.

[174] SACCO R L, DEROSA J T, HALEY E C, JR., et al. Glycine antagonist in

neuroprotection for patients with acute stroke: GAIN Americas: a randomized

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 206

controlled trial. Jama, 2001, 285(13): 1719-1728.

[175] DIENER H C, HACKE W, HENNERICI M, et al. Lubeluzole in acute ischemic

stroke. A double-blind, placebo-controlled phase II trial. Lubeluzole International

Study Group. Stroke, 1996, 27(1): 76-81.

[176] DIENER H C, CORTENS M, FORD G, et al. Lubeluzole in acute ischemic

stroke treatment: A double-blind study with an 8-hour inclusion window comparing a

10-mg daily dose of lubeluzole with placebo. Stroke, 2000, 31(11): 2543-2551.

[177] GROTTA J. Lubeluzole treatment of acute ischemic stroke. The US and

Canadian Lubeluzole Ischemic Stroke Study Group. Stroke, 1997, 28(12): 2338-2346.

[178] LEES K R, SHARMA A K, BARER D, et al. Tolerability and pharmacokinetics

of the nitrone NXY-059 in patients with acute stroke. Stroke, 2001, 32(3): 675-680.

[179] LEES K R, ZIVIN J A, ASHWOOD T, et al. NXY-059 for acute ischemic

stroke. N Engl J Med, 2006, 354(6): 588-600.

[180] SHUAIB A, LEES K R, LYDEN P, et al. NXY-059 for the treatment of acute

ischemic stroke. N Engl J Med, 2007, 357(6): 562-571.

[181] MUIR K W, LEES K R. A randomized, double-blind, placebo-controlled pilot

trial of intravenous magnesium sulfate in acute stroke. Stroke, 1995, 26(7): 1183-

1188.

[182] LAMPL Y, GILAD R, GEVA D, et al. Intravenous administration of magnesium

sulfate in acute stroke: a randomized double-blind study. Clin Neuropharmacol, 2001,

24(1): 11-15.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 207

[183] MUIR K W, LEES K R, FORD I, et al. Magnesium for acute stroke

(Intravenous Magnesium Efficacy in Stroke trial): randomised controlled trial. Lancet,

2004, 363(9407): 439-445.

[184] ZHAO L, ZHAO C Y. Observing the effect of magnesium sulfate in patients

with acute ischemia stroke. Shanxi Clinical Medicine, 2000, 9(12): 901-902.

[185] ZHU P H, DENG T X. Clinical efficacy of intravenous magnesium sulfate in

patients with acute ischemic stroke. China Journal of Pharmaceutical Economics,

2014, 7): 46-47.

[186] SAVER J L, KIDWELL C, ECKSTEIN M, et al. Prehospital neuroprotective

therapy for acute stroke: results of the Field Administration of Stroke Therapy-

Magnesium (FAST-MAG) pilot trial. Stroke, 2004, 35(5): e106-108.

[187] SAVER J L, STARKMAN S, ECKSTEIN M, et al. Prehospital use of

magnesium sulfate as neuroprotection in acute stroke. N Engl J Med, 2015, 372(6):

528-536.

[188] CLARK W M, WARACH S J, PETTIGREW L C, et al. A randomized dose-

response trial of citicoline in acute ischemic stroke patients. Citicoline Stroke Study

Group. Neurology, 1997, 49(3): 671-678.

[189] CLARK W M, WECHSLER L R, SABOUNJIAN L A, et al. A phase III

randomized efficacy trial of 2000 mg citicoline in acute ischemic stroke patients.

Neurology, 2001, 57(9): 1595-1602.

[190] CLARK W M, WILLIAMS B J, SELZER K A, et al. A randomized efficacy

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 208

trial of citicoline in patients with acute ischemic stroke. Stroke, 1999, 30(12): 2592-

2597.

[191] SAVER J L. Citicoline: update on a promising and widely available agent for

neuroprotection and neurorepair. Reviews in neurological diseases, 2008, 5(4): 167-

177.

[192] DAVALOS A, CASTILLO J, ALVAREZ-SABIN J, et al. Oral citicoline in acute

ischemic stroke: an individual patient data pooling analysis of clinical trials. Stroke,

2002, 33(12): 2850-2857.

[193] Ganglioside GM1 in acute ischemic stroke. The SASS Trial. Stroke, 1994,

25(6): 1141-1148.

[194] LENZI G L, GRIGOLETTO F, GENT M, et al. Early treatment of stroke with

monosialoganglioside GM-1. Efficacy and safety results of the Early Stroke Trial.

Stroke, 1994, 25(8): 1552-1558.

[195] CANDELISE L, CICCONE A. Gangliosides for acute ischaemic stroke.

Cochrane Database Syst Rev, 2000, 2): Cd000094.

[196] ZHANG L H, LI D M. Effect of Argatroban + Brain Protective Agentin in the

Treatment of Acute Ischemic Stroke. Chinese Journal of Medical Guide, 2017, 06):

586-587.

[197] SU Y, FAN L, ZHANG Y, et al. Improved Neurological Outcome With Mild

Hypothermia in Surviving Patients With Massive Cerebral Hemispheric Infarction.

Stroke, 2016, 47(2): 457-463.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 209

[198] PIIRONEN K, TIAINEN M, MUSTANOJA S, et al. Mild hypothermia after

intravenous thrombolysis in patients with acute stroke: a randomized controlled trial.

Stroke, 2014, 45(2): 486-491.

[199] HEMMEN T M, RAMAN R, GULUMA K Z, et al. Intravenous thrombolysis

plus hypothermia for acute treatment of ischemic stroke (ICTuS-L): final results.

Stroke, 2010, 41(10): 2265-2270.

[200] LYDEN P, HEMMEN T, GROTTA J, et al. Results of the ICTuS 2 Trial

(Intravascular Cooling in the Treatment of Stroke 2). Stroke, 2016, 47(12): 2888-

2895.

[201] HORN C M, SUN C H, NOGUEIRA R G, et al. Endovascular Reperfusion and

Cooling in Cerebral Acute Ischemia (ReCCLAIM I). J Neurointerv Surg, 2014, 6(2):

91-95.

[202] HONG J M, LEE J S, SONG H J, et al. Therapeutic hypothermia after

recanalization in patients with acute ischemic stroke. Stroke, 2014, 45(1): 134-140.

[203] OVESEN C, BRIZZI M, POTT F C, et al. Feasibility of endovascular and

surface cooling strategies in acute stroke. Acta Neurol Scand, 2013, 127(6): 399-405.

[204] GEURTS M, PETERSSON J, BRIZZI M, et al. COOLIST (Cooling for

Ischemic Stroke Trial): A Multicenter, Open, Randomized, Phase II, Clinical Trial.

Stroke, 2017, 48(1): 219-221.

[205] SCHWAB S, GEORGIADIS D, BERROUSCHOT J, et al. Feasibility and

safety of moderate hypothermia after massive hemispheric infarction. Stroke, 2001,

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 210

32(9): 2033-2035.

[206] JIAN S, YONGMING Q, ZHIHUA C, et al. Feasibility and safety of moderate

hypothermia after acute ischemic stroke. International journal of developmental

neuroscience : the official journal of the International Society for Developmental

Neuroscience, 2003, 21(6): 353-356.

[207] CHEN C H, CHEN S Y, WANG V, et al. Effects of repetitive hyperbaric oxygen

treatment in patients with acute cerebral infarction: a pilot study.

TheScientificWorldJournal, 2012, 2012(694703.

[208] Hu Y X. Effects of early hyperbaric oxygen combined with joint training on

motor function in patients with acute ischemic stroke. Central Plains Medical Journal,

2014, 22): 35-36.

[209] Ren X S, YANG B. Randomized controlled study of hyperbaric oxygen in the

treatment of acute ischemic stroke. Chinese Journal of Health Care Nutrition, 2016,

25): 153-153.

[210] CAO C Y. Hyperbaric oxygen for acute ischemic stroke:a systematic review of

randomized controlled trials. MEDICAL JOURNAL OF THE CHINESE PEOPLE'S

ARMED POLICE FORCES, 2002, 2): 112-116.

[211] PENG H L. A systematic review of randomized controlled trials of hyperbaric

oxygen in the treatment of acute ischemic stroke. Medical Frontier, 2017, 3): 372-373.

[212] BENNETT M H, WEIBEL S, WASIAK J, et al. Hyperbaric oxygen therapy for

acute ischaemic stroke. Cochrane Database Syst Rev, 2014, 11): Cd004954.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 211

[213] HEYBOER M, 3RD, SHARMA D, SANTIAGO W, et al. Hyperbaric Oxygen

Therapy: Side Effects Defined and Quantified. Advances in wound care, 2017, 6(6):

210-224.

[214] HEYBOER M, 3RD, JENNINGS S, GRANT W D, et al. Seizure incidence by

treatment pressure in patients undergoing hyperbaric oxygen therapy. Undersea &

hyperbaric medicine : journal of the Undersea and Hyperbaric Medical Society, Inc,

2014, 41(5): 379-385.

[215] TEKLE W G, ADKINSON C D, CHAUDHRY S A, et al. Factors associated

with favorable response to hyperbaric oxygen therapy among patients presenting with

iatrogenic cerebral arterial gas embolism. Neurocritical care, 2013, 18(2): 228-233.

[216] WEIXLER V H, YATES A E, PUCHINGER M, et al. Hyperbaric oxygen in

patients with ischemic stroke following cardiac surgery: a retrospective observational

trial. Undersea & hyperbaric medicine : journal of the Undersea and Hyperbaric

Medical Society, Inc, 2017, 44(5): 377-385.

[217] APPLEBAUM R M, KASLIWAL R, TUNICK P A, et al. Sequential external

counterpulsation increases cerebral and renal blood flow. American heart journal,

1997, 133(6): 611-615.

[218] HAN J H, LEUNG T W, LAM W W, et al. Preliminary findings of external

counterpulsation for ischemic stroke patient with large artery occlusive disease.

Stroke, 2008, 39(4): 1340-1343.

[219] GULUMA K Z, LIEBESKIND D S, RAMAN R, et al. Feasibility and Safety of

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 212

Using External Counterpulsation to Augment Cerebral Blood Flow in Acute Ischemic

Stroke-The Counterpulsation to Upgrade Forward Flow in Stroke (CUFFS) Trial.

Journal of stroke and cerebrovascular diseases : the official journal of National Stroke

Association, 2015, 24(11): 2596-2604.

[220] SHUAIB A, BORNSTEIN N M, DIENER H C, et al. Partial aortic occlusion

for cerebral perfusion augmentation: safety and efficacy of NeuroFlo in Acute

Ischemic Stroke trial. Stroke, 2011, 42(6): 1680-1690.

[221] HAMMER M D, SCHWAMM L, STARKMAN S, et al. Safety and feasibility

of NeuroFlo use in eight- to 24-hour ischemic stroke patients. International journal of

stroke : official journal of the International Stroke Society, 2012, 7(8): 655-661.

[222] MEIER F, WESSEL G, THIELE R, et al. Induced hypertension as an approach

to treating acute cerebrovascular ischaemia: possibilities and limitations.

Experimental pathology, 1991, 42(4): 257-263.

[223] STRAND T, ASPLUND K, ERIKSSON S, et al. A randomized controlled trial

of hemodilution therapy in acute ischemic stroke. Stroke, 1984, 15(6): 980-989.

[224] ASPLUND K. Randomized clinical trials of hemodilution in acute ischemic

stroke. Acta neurologica Scandinavica Supplementum, 1989, 127(22-30.

[225] GOSLINGA H, HEUVELMANS J H, SCHMID-SCHONBEIN H.

Hemodilution and rehydration in acute ischemic stroke. A preliminary report on the

Amsterdam Stroke Study. Acta medica Austriaca, 1991, 18 Suppl 1(41-44.

[226] GOSLINGA H, EIJZENBACH V, HEUVELMANS J H, et al. Custom-tailored

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 213

hemodilution with albumin and crystalloids in acute ischemic stroke. Stroke, 1992,

23(2): 181-188.

[227] STRAND T. Evaluation of long-term outcome and safety after hemodilution

therapy in acute ischemic stroke. Stroke, 1992, 23(5): 657-662.

[228] CHANG T S, JENSEN M B. Haemodilution for acute ischaemic stroke.

Cochrane Database Syst Rev, 2014, 8): Cd000103.

[229] MILLER J B, LEWANDOWSKI C, WIRA C R, et al. Volume of Plasma

Expansion and Functional Outcomes in Stroke. Neurocritical care, 2017, 26(2): 191-

195.

[230] LAMPL Y, ZIVIN J A, FISHER M, et al. Infrared laser therapy for ischemic

stroke: a new treatment strategy: results of the NeuroThera Effectiveness and Safety

Trial-1 (NEST-1). Stroke, 2007, 38(6): 1843-1849.

[231] ZIVIN J A, ALBERS G W, BORNSTEIN N, et al. Effectiveness and safety of

transcranial laser therapy for acute ischemic stroke. Stroke, 2009, 40(4): 1359-1364.

[232] HUISA B N, STEMER A B, WALKER M G, et al. Transcranial laser therapy

for acute ischemic stroke: a pooled analysis of NEST-1 and NEST-2. International

journal of stroke : official journal of the International Stroke Society, 2013, 8(5): 315-

320.

[233] KASNER S E, ROSE D Z, SKOKAN A, et al. Transcranial laser therapy and

infarct volume. Stroke, 2013, 44(7): 2025-2027.

[234] HACKE W, SCHELLINGER P D, ALBERS G W, et al. Transcranial laser

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 214

therapy in acute stroke treatment: results of neurothera effectiveness and safety trial 3,

a phase III clinical end point device trial. Stroke, 2014, 45(11): 3187-3193.

[235] ZIVIN J A, SEHRA R, SHOSHOO A, et al. NeuroThera(R) Efficacy and Safety

Trial-3 (NEST-3): a double-blind, randomized, sham-controlled, parallel group,

multicenter, pivotal study to assess the safety and efficacy of transcranial laser therapy

with the NeuroThera(R) Laser System for the treatment of acute ischemic stroke

within 24 h of stroke onset. International journal of stroke : official journal of the

International Stroke Society, 2014, 9(7): 950-955.

[236] DZIEDZIC T, SLOWIK A, SZCZUDLIK A. Serum albumin level as a predictor

of ischemic stroke outcome. Stroke, 2004, 35(6): e156-158.

[237] CHO Y M, CHOI I S, BIAN R X, et al. Serum albumin at admission for

prediction of functional outcome in ischaemic stroke patients. Neurological sciences :

official journal of the Italian Neurological Society and of the Italian Society of

Clinical Neurophysiology, 2008, 29(6): 445-449.

[238] SUN F H, LI S J, LIU Y Y, et al. Observation of the Effect of Human

Albumins in Acute Severe Cerebral Infarction Patients. Chinese Journal of Stroke,

2009, 12): 951-955.

[239] GINSBERG M D, PALESCH Y Y, MARTIN R H, et al. The albumin in acute

stroke (ALIAS) multicenter clinical trial: safety analysis of part 1 and rationale and

design of part 2. Stroke, 2011, 42(1): 119-127.

[240] HILL M D, MARTIN R H, PALESCH Y Y, et al. The Albumin in Acute Stroke

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 215

Part 1 Trial: an exploratory efficacy analysis. Stroke, 2011, 42(6): 1621-1625.

[241] GINSBERG M D, HILL M D, PALESCH Y Y, et al. The ALIAS Pilot Trial: a

dose-escalation and safety study of albumin therapy for acute ischemic stroke--I:

Physiological responses and safety results. Stroke, 2006, 37(8): 2100-2106.

[242] PALESCH Y Y, HILL M D, RYCKBORST K J, et al. The ALIAS Pilot Trial: a

dose-escalation and safety study of albumin therapy for acute ischemic stroke--II:

neurologic outcome and efficacy analysis. Stroke, 2006, 37(8): 2107-2114.

[243] KOCH S, CONCHA M, WAZZAN T, et al. High dose human serum albumin

for the treatment of acute ischemic stroke: a safety study. Neurocritical care, 2004,

1(3): 335-341.

[244] HILL M D, MARTIN R H, PALESCH Y Y, et al. Albumin Administration in

Acute Ischemic Stroke: Safety Analysis of the ALIAS Part 2 Multicenter Trial. PLoS

One, 2015, 10(9): e0131390.

[245] MARTIN R H, YEATTS S D, HILL M D, et al. ALIAS (Albumin in Acute

Ischemic Stroke) Trials: Analysis of the Combined Data From Parts 1 and 2. Stroke,

2016, 47(9): 2355-2359.

[246] KHATRI R, AFZAL M R, RODRIGUEZ G J, et al. Albumin-Induced

Neuroprotection in Focal Cerebral Ischemia in the ALIAS Trial: Does Severity,

Mechanism, and Time of Infusion Matter? Neurocritical care, 2018, 28(1): 60-64.

[247] SULTER G, ELTING J W, STEWART R, et al. Continuous pulse oximetry in

acute hemiparetic stroke. J Neurol Sci, 2000, 179(S 1-2): 65-69.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 216

[248] ROFFE C, SILLS S, HALIM M, et al. Unexpected nocturnal hypoxia in

patients with acute stroke. Stroke, 2003, 34(11): 2641-2645.

[249] AVIV J E, MARTIN J H, SACCO R L, et al. Supraglottic and pharyngeal

sensory abnormalities in stroke patients with dysphagia. Ann Otol Rhinol Laryngol,

1996, 105(2): 92-97.

[250] MILHAUD D, POPP J, THOUVENOT E, et al. Mechanical ventilation in

ischemic stroke. Journal of stroke and cerebrovascular diseases : the official journal of

National Stroke Association, 2004, 13(4): 183-188.

[251] ROFFE C, NEVATTE T, SIM J, et al. Effect of Routine Low-Dose Oxygen

Supplementation on Death and Disability in Adults With Acute Stroke: The Stroke

Oxygen Study Randomized Clinical Trial. Jama, 2017, 318(12): 1125-1135.

[252] ALI K, WARUSEVITANE A, LALLY F, et al. The stroke oxygen pilot study: a

randomized controlled trial of the effects of routine oxygen supplementation early

after acute stroke--effect on key outcomes at six months. PLoS One, 2014, 8(6):

e59274.

[253] SINGHAL A, INVESTIGATORS P S. A phase IIB clinical trial of normobaric

oxygen therapy (NBO) in acute ischemic stroke (AIS). Neurology, 2013, 80(suppl):

S02.001.

[254] ROFFE C, ALI K, WARUSEVITANE A, et al. The SOS pilot study: a RCT of

routine oxygen supplementation early after acute stroke--effect on recovery of

neurological function at one week. PLoS One, 2011, 6(5): e19113.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 217

[255] ROFFE C, SILLS S, POUNTAIN S J, et al. A randomized controlled trial of the

effect of fixed-dose routine nocturnal oxygen supplementation on oxygen saturation

in patients with acute stroke. Journal of stroke and cerebrovascular diseases : the

official journal of National Stroke Association, 2010, 19(1): 29-35.

[256] SINGHAL A B, BENNER T, ROCCATAGLIATA L, et al. A pilot study of

normobaric oxygen therapy in acute ischemic stroke. Stroke, 2005, 36(4): 797-802.

[257] RONNING O M, GULDVOG B. Should stroke victims routinely receive

supplemental oxygen? A quasi-randomized controlled trial. Stroke, 1999, 30(10):

2033-2037.

[258] DOGGETT D L, TAPPE K A, MITCHELL M D, et al. Prevention of

pneumonia in elderly stroke patients by systematic diagnosis and treatment of

dysphagia: an evidence-based comprehensive analysis of the literature. Dysphagia,

2001, 16(4): 279-295.

[259] RANGEL-CASTILLA L, GOPINATH S, ROBERTSON C S. Management of

intracranial hypertension. Neurol Clin, 2008, 26(2): 521-541, x.

[260] GROTTA J, PASTEUR W, KHWAJA G, et al. Elective intubation for

neurologic deterioration after stroke. Neurology, 1995, 45(4): 640-644.

[261] FOERCH C, KESSLER K R, STECKEL D A, et al. Survival and quality of life

outcome after mechanical ventilation in elderly stroke patients. Journal of neurology,

neurosurgery, and psychiatry, 2004, 75(7): 988-993.

[262] BUSHNELL C D, PHILLIPS-BUTE B G, LASKOWITZ D T, et al. Survival

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 218

and outcome after endotracheal intubation for acute stroke. Neurology, 1999, 52(7):

1374-1381.

[263] HOLLOWAY R G, BENESCH C G, BURGIN W S, et al. Prognosis and

decision making in severe stroke. Jama, 2005, 294(6): 725-733.

[264] GOLESTANIAN E, LIOU J I, SMITH M A. Long-term survival in older

critically ill patients with acute ischemic stroke. Crit Care Med, 2009, 37(12): 3107-

3113.

[265] AZZIMONDI G, BASSEIN L, NONINO F, et al. Fever in acute stroke worsens

prognosis. A prospective study. Stroke, 1995, 26(11): 2040-2043.

[266] JORGENSEN H S, REITH J, NAKAYAMA H, et al. What determines good

recovery in patients with the most severe strokes? The Copenhagen Stroke Study.

Stroke, 1999, 30(10): 2008-2012.

[267] DEN HERTOG H M, VAN DER WORP H B, VAN GEMERT H M, et al. The

Paracetamol (Acetaminophen) In Stroke (PAIS) trial: a multicentre, randomised,

placebo-controlled, phase III trial. The Lancet Neurology, 2009, 8(5): 434-440.

[268] DEN HERTOG H M, VAN DER WORP H B, VAN GEMERT H M, et al. An

early rise in body temperature is related to unfavorable outcome after stroke: data

from the PAIS study. Journal of neurology, 2011, 258(2): 302-307.

[269] SAXENA M, YOUNG P, PILCHER D, et al. Early temperature and mortality in

critically ill patients with acute neurological diseases: trauma and stroke differ from

infection. Intensive Care Med, 2015, 41(5): 823-832.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 219

[270] LYDEN P, HEMMEN T, GROTTA J, et al. Results of the ICTuS 2 Trial

(Intravascular Cooling in the Treatment of Stroke 2). Stroke, 2016, 47(12): 2888-

2895.

[271] CHOI-KWON S, YANG Y H, KIM E K, et al. Nutritional status in acute stroke:

undernutrition versus overnutrition in different stroke subtypes. Acta Neurol Scand,

1998, 98(3): 187-192.

[272] GARIBALLA S E, PARKER S G, TAUB N, et al. Influence of nutritional status

on clinical outcome after acute stroke. Am J Clin Nutr, 1998, 68(2): 275-281.

[273] DENNIS M, LEWIS S, CRANSWICK G, et al. FOOD: a multicentre

randomised trial evaluating feeding policies in patients admitted to hospital with a

recent stroke. Health Technol Assess, 2006, 10(2): iii-iv, ix-x, 1-120.

[274] GEEGANAGE C, BEAVAN J, ELLENDER S, et al. Interventions for

dysphagia and nutritional support in acute and subacute stroke. Cochrane Database

Syst Rev, 2012, 10(CD000323.

[275] SORENSEN R T, RASMUSSEN R S, OVERGAARD K, et al. Dysphagia

screening and intensified oral hygiene reduce pneumonia after stroke. J Neurosci

Nurs, 2013, 45(3): 139-146.

[276] BRADY M, FURLANETTO D, HUNTER R V, et al. Staff-led interventions for

improving oral hygiene in patients following stroke. Cochrane Database Syst Rev,

2006, 4): CD003864.

[277] WAGNER C, MARCHINA S, DEVEAU J A, et al. Risk of Stroke-Associated

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 220

Pneumonia and Oral Hygiene. Cerebrovasc Dis, 2016, 41(1-2): 35-39.

[278] VAN DER WORP H B, KAPPELLE L J. Complications of acute ischaemic

stroke. Cerebrovasc Dis, 1998, 8(2): 124-132.

[279] MARTINO R, FOLEY N, BHOGAL S, et al. Dysphagia after stroke: incidence,

diagnosis, and pulmonary complications. Stroke, 2005, 36(12): 2756-2763.

[280] ASLANYAN S, WEIR C J, DIENER H C, et al. Pneumonia and urinary tract

infection after acute ischaemic stroke: a tertiary analysis of the GAIN International

trial. Eur J Neurol, 2004, 11(1): 49-53.

[281] FIELD T S, GREEN T L, ROY K, et al. Trends in hospital admission for stroke

in Calgary. Can J Neurol Sci, 2004, 31(3): 387-393.

[282] NAKAGAWA T, SEKIZAWA K, ARAI H, et al. High incidence of pneumonia

in elderly patients with basal ganglia infarction. Archives of internal medicine, 1997,

157(3): 321-324.

[283] HILKER R, POETTER C, FINDEISEN N, et al. Nosocomial pneumonia after

acute stroke: implications for neurological intensive care medicine. Stroke, 2003,

34(4): 975-981.

[284] UPADYA A, THOREVSKA N, SENA K N, et al. Predictors and consequences

of pneumonia in critically ill patients with stroke. J Crit Care, 2004, 19(1): 16-22.

[285] CHAMORRO A, HORCAJADA J P, OBACH V, et al. The Early Systemic

Prophylaxis of Infection After Stroke study: a randomized clinical trial. Stroke, 2005,

36(7): 1495-1500.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 221

[286] LANGHORNE P, STOTT D J, ROBERTSON L, et al. Medical complications

after stroke: a multicenter study. Stroke, 2000, 31(6): 1223-1229.

[287] ROTH E J, LOVELL L, HARVEY R L, et al. Incidence of and risk factors for

medical complications during stroke rehabilitation. Stroke, 2001, 32(2): 523-529.

[288] KONG K H, YOUNG S. Incidence and outcome of poststroke urinary retention:

a prospective study. Arch Phys Med Rehabil, 2000, 81(11): 1464-1467.

[289] MCLEAN D E. Medical complications experienced by a cohort of stroke

survivors during inpatient, tertiary-level stroke rehabilitation. Arch Phys Med

Rehabil, 2004, 85(3): 466-469.

[290] WEEN J E, ALEXANDER M P, D'ESPOSITO M, et al. Incontinence after

stroke in a rehabilitation setting: outcome associations and predictive factors.

Neurology, 1996, 47(3): 659-663.

[291] JAUCH E C, SAVER J L, ADAMS H P, JR., et al. Guidelines for the early

management of patients with acute ischemic stroke: a guideline for healthcare

professionals from the American Heart Association/American Stroke Association.

Stroke, 2013, 44(3): 870-947.

[292] COLLABORATION C T, DENNIS M, SANDERCOCK P, et al. Effectiveness

of intermittent pneumatic compression in reduction of risk of deep vein thrombosis in

patients who have had a stroke (CLOTS 3): a multicentre randomised controlled trial.

Lancet, 2013, 382(9891): 516-524.

[293] DENNIS M, CASO V, KAPPELLE L J, et al. European Stroke Organisation

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 222

(ESO) guidelines for prophylaxis for venous thromboembolism in immobile patients

with acute ischaemic stroke. Eur Stroke J, 2016, 1(1): 6-19.

[294] WHITELEY W N, ADAMS H P, JR., BATH P M, et al. Targeted use of heparin,

, or low-molecular-weight heparin to improve outcome after acute

ischaemic stroke: an individual patient data meta-analysis of randomised controlled

trials. The Lancet Neurology, 2013, 12(6): 539-545.

[295] SANDERCOCK P A, COUNSELL C, KANE E J. Anticoagulants for acute

ischaemic stroke. Cochrane Database Syst Rev, 2015, 3): CD000024.

[296] GROUP A T C. Efficacy and safety of very early mobilisation within 24 h of

stroke onset (AVERT): a randomised controlled trial. Lancet, 2015, 386(9988): 46-55.

[297] EUROPEAN STROKE INITIATIVE WRITING C, WRITING COMMITTEE

FOR THE E E C, STEINER T, et al. Recommendations for the management of

intracranial haemorrhage - part I: spontaneous intracerebral haemorrhage. The

European Stroke Initiative Writing Committee and the Writing Committee for the

EUSI Executive Committee. Cerebrovasc Dis, 2006, 22(4): 294-316.

[298] WAGNER I, HAUER E M, STAYKOV D, et al. Effects of continuous

hypertonic saline infusion on perihaemorrhagic oedema evolution. Stroke, 2011,

42(6): 1540-1545.

[299] KOENIG M A, BRYAN M, LEWIN J L, 3RD, et al. Reversal of transtentorial

herniation with hypertonic saline. Neurology, 2008, 70(13): 1023-1029.

[300] CURLEY G, KAVANAGH B P, LAFFEY J G. Hypocapnia and the injured

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 223

brain: more harm than benefit. Crit Care Med, 2010, 38(5): 1348-1359.

[301] WAN Y H, NIE C, WANG H L, et al. Therapeutic hypothermia (different

depths, durations, and rewarming speeds) for acute ischemic stroke: a meta-analysis.

Journal of stroke and cerebrovascular diseases : the official journal of National Stroke

Association, 2014, 23(10): 2736-2747.

[302] MANNO E M, NICHOLS D A, FULGHAM J R, et al. Computed tomographic

determinants of neurologic deterioration in patients with large middle cerebral artery

infarctions. Mayo Clin Proc, 2003, 78(2): 156-160.

[303] HEINSIUS T, BOGOUSSLAVSKY J, VAN MELLE G. Large infarcts in the

middle cerebral artery territory. Aetiology and outcome patterns. Neurology, 1998,

50(2): 341-350.

[304] WIJDICKS E F, DIRINGER M N. Middle cerebral artery territory infarction

and early brain swelling: progression and effect of age on outcome. Mayo Clin Proc,

1998, 73(9): 829-836.

[305] THOMALLA G J, KUCINSKI T, SCHODER V, et al. Prediction of malignant

middle cerebral artery infarction by early perfusion- and diffusion-weighted magnetic

resonance imaging. Stroke, 2003, 34(8): 1892-1899.

[306] CHO D Y, CHEN T C, LEE H C. Ultra-early decompressive craniectomy for

malignant middle cerebral artery infarction. Surg Neurol, 2003, 60(3): 227-232;

discussion 232-223.

[307] RYOO J W, NA D G, KIM S S, et al. Malignant middle cerebral artery

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 224

infarction in hyperacute ischemic stroke: evaluation with multiphasic perfusion

computed tomography maps. J Comput Assist Tomogr, 2004, 28(1): 55-62.

[308] QURESHI A I, SUAREZ J I, YAHIA A M, et al. Timing of neurologic

deterioration in massive middle cerebral artery infarction: a multicenter review. Crit

Care Med, 2003, 31(1): 272-277.

[309] ROPPER A H, SHAFRAN B. Brain oedema after stroke. Clinical syndrome and

intracranial pressure. Arch Neurol, 1984, 41(1): 26-29.

[310] TONI D, FIORELLI M, GENTILE M, et al. Progressing neurological deficit

secondary to acute ischemic stroke. A study on predictability, pathogenesis, and

prognosis. Arch Neurol, 1995, 52(7): 670-675.

[311] ABE M, UDONO H, TABUCHI K, et al. Analysis of ischemic brain damage in

cases of acute subdural hematomas. Surg Neurol, 2003, 59(6): 464-472; discussion

472.

[312] HUSSAIN S I, CORDERO-TUMANGDAY C, GOLDENBERG F D, et al.

Brainstem ischemia in acute herniation syndrome. J Neurol Sci, 2008, 268(1-2): 190-

192.

[313] VAHEDI K, HOFMEIJER J, JUETTLER E, et al. Early decompressive surgery

in malignant infarction of the middle cerebral artery: a pooled analysis of three

randomised controlled trials. The Lancet Neurology, 2007, 6(3): 215-222.

[314] MACIEL C B, SHETH K N. Malignant MCA Stroke: an Update on Surgical

Decompression and Future Directions. Curr Atheroscler Rep, 2015, 17(7): 40.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 225

[315] YANG M H, LIN H Y, FU J, et al. Decompressive hemicraniectomy in patients

with malignant middle cerebral artery infarction: A systematic review and meta-

analysis. Surgeon, 2015, 13(4): 230-240.

[316] SUNDSETH J, SUNDSETH A, JACOBSEN E A, et al. Predictors of early in-

hospital death after decompressive craniectomy in swollen middle cerebral artery

infarction. Acta Neurochir (Wien), 2017, 159(2): 301-306.

[317] SUYAMA K, HORIE N, HAYASHI K, et al. Nationwide survey of

decompressive hemicraniectomy for malignant middle cerebral artery infarction in

Japan. World neurosurgery, 2014, 82(6): 1158-1163.

[318] YU J W, CHOI J H, KIM D H, et al. Outcome following decompressive

craniectomy for malignant middle cerebral artery infarction in patients older than 70

years old. J Cerebrovasc Endovasc Neurosurg, 2012, 14(2): 65-74.

[319] JUTTLER E, UNTERBERG A, WOITZIK J, et al. Hemicraniectomy in older

patients with extensive middle-cerebral-artery stroke. N Engl J Med, 2014, 370(12):

1091-1100.

[320] ZHAO J, SU Y Y, ZHANG Y, et al. Decompressive hemicraniectomy in

malignant middle cerebral artery infarct: a randomized controlled trial enrolling

patients up to 80 years old. Neurocritical care, 2012, 17(2): 161-171.

[321] RACO A, CAROLI E, ISIDORI A, et al. Management of acute cerebellar

infarction: one institution's experience. Neurosurgery, 2003, 53(5): 1061-1065;

discussion 1065-1066.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 226

[322] AGARWALLA P K, STAPLETON C J, OGILVY C S. Craniectomy in acute

ischemic stroke. Neurosurgery, 2014, 74 Suppl 1(S151-162.

[323] MOSTOFI K. Neurosurgical management of massive cerebellar infarct outcome

in 53 patients. Surg Neurol Int, 2013, 4(28.

[324] MOTTO C, ARITZU E, BOCCARDI E, et al. Reliability of haemorrhagic

transformation diagnosis in acute ischemic stroke. Stroke, 1997, 28(2): 302-306.

[325] MOTTO C, CICCONE A, ARITZU E, et al. Haemorrhage after an acute

ischemic stroke.MAST-I Collaborative Group. Stroke, 1999, 30(4): 761-764.

[326] DEREX L, HERMIER M, ADELEINE P, et al. Clinical and imaging predictors

of intracerebral haemorrhage in stroke patients treated with intravenous tissue

plasminogen activator. Journal of neurology, neurosurgery, and psychiatry, 2005,

76(1): 70-75.

[327] LEIGH R, ZAIDAT O O, SURI M F, et al. Predictors of hyperacute clinical

worsening in ischemic stroke patients receiving thrombolytic therapy. Stroke, 2004,

35(8): 1903-1907.

[328] BOGOUSSLAVSKY J, REGLI F. Anticoagulant-induced intracerebral bleeding

in brain ischemia. Evaluation in 200 patients with TIAs, emboli from the heart, and

progressing stroke. Acta Neurol Scand, 1985, 71(6): 464-471.

[329] WARACH S, LATOUR L L. Evidence of reperfusion injury, exacerbated by

thrombolytic therapy, in human focal brain ischemia using a novel imaging marker of

early blood-brain barrier disruption. Stroke, 2004, 35(11 Suppl 1): 2659-2661.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 227

[330] Intracerebral haemorrhage after intravenous t-PA therapy for ischemic stroke.

The NINDS t-PA Stroke Study Group. Stroke, 1997, 28(11): 2109-2118.

[331] YAGHI S, BOEHME A K, DIBU J, et al. Treatment and Outcome of

Thrombolysis-Related Haemorrhage: A Multicenter Retrospective Study. JAMA

Neurol, 2015, 72(12): 1451-1457.

[332] KIM J T, HEO S H, PARK M S, et al. Use of after

haemorrhagic transformation in acute ischemic stroke. PLoS One, 2014, 9(2): e89798.

[333] ENGLAND T J, BATH P M, SARE G M, et al. Asymptomatic haemorrhagic

transformation of infarction and its relationship with functional outcome and stroke

subtype: assessment from the Tinzaparin in Acute Ischaemic Stroke Trial. Stroke,

2010, 41(12): 2834-2839.

[334] KILINCER C, ASIL T, UTKU U, et al. Factors affecting the outcome of

decompressive craniectomy for large hemispheric infarctions: a prospective cohort

study. Acta Neurochir (Wien), 2005, 147(6): 587-594; discussion 594.

[335] BURN J, DENNIS M, BAMFORD J, et al. Epileptic seizures after a first

stroke: the Oxfordshire Community Stroke Project. BMJ, 1997, 315(7122): 1582-

1587.

[336] ALBERTI A, PACIARONI M, CASO V, et al. Early seizures in patients with

acute stroke: frequency, predictive factors, and effect on clinical outcome. Vascular

health and risk management, 2008, 4(3): 715-720.

[337] AWADA A, OMOJOLA M F, OBEID T. Late epileptic seizures after cerebral

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 228

infarction. Acta Neurol Scand, 1999, 99(5): 265-268.

[338] CAMILO O, GOLDSTEIN L B. Seizures and epilepsy after ischemic stroke.

Stroke, 2004, 35(7): 1769-1775.

[339] WARDLAW J M, SEYMOUR J, CAIRNS J, et al. Immediate computed

tomography scanning of acute stroke is cost-effective and improves quality of life.

Stroke; a journal of cerebral circulation, 2004, 35(11): 2477-2483.

[340] BARBER P A, HILL M D, ELIASZIW M, et al. Imaging of the brain in acute

ischaemic stroke: comparison of computed tomography and magnetic resonance

diffusion-weighted imaging. Journal of neurology, neurosurgery, and psychiatry,

2005, 76(11): 1528-1533.

[341] CHALELA J A, KIDWELL C S, NENTWICH L M, et al. Magnetic resonance

imaging and computed tomography in emergency assessment of patients with

suspected acute stroke: a prospective comparison. Lancet (London, England), 2007,

369(9558): 293-298.

[342] HEIDENREICH J O, HSU D, WANG G, et al. Magnetic resonance imaging

results can affect therapy decisions in hyperacute stroke care. Acta radiologica

(Stockholm, Sweden : 1987), 2008, 49(5): 550-557.

[343] HWANG D Y, SILVA G S, FURIE K L, et al. Comparative sensitivity of

computed tomography vs. magnetic resonance imaging for detecting acute posterior

fossa infarct. The Journal of emergency medicine, 2012, 42(5): 559-565.

[344] WARDLAW J, BRAZZELLI M, MIRANDA H, et al. An assessment of the

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 229

cost-effectiveness of magnetic resonance, including diffusion-weighted imaging, in

patients with transient ischaemic attack and minor stroke: a systematic review, meta-

analysis and economic evaluation. Health technology assessment (Winchester,

England), 2014, 18(27): 1-368, v-vi.

[345] PATEL S C, LEVINE S R, TILLEY B C, et al. Lack of clinical significance of

early ischemic changes on computed tomography in acute stroke. Jama, 2001,

286(22): 2830-2838.

[346] DEMCHUK A M, HILL M D, BARBER P A, et al. Importance of early

ischemic computed tomography changes using ASPECTS in NINDS rtPA Stroke

Study. Stroke; a journal of cerebral circulation, 2005, 36(10): 2110-2115.

[347] DEMCHUK A M, KHAN F, HILL M D, et al. Importance of leukoaraiosis on

CT for tissue plasminogen activator decision making: evaluation of the NINDS rt-PA

Stroke Study. Cerebrovasc Dis, 2008, 26(2): 120-125.

[348] DZIALOWSKI I, HILL M D, COUTTS S B, et al. Extent of early ischemic

changes on computed tomography (CT) before thrombolysis: prognostic value of the

Alberta Stroke Program Early CT Score in ECASS II. Stroke; a journal of cerebral

circulation, 2006, 37(4): 973-978.

[349] Association between brain imaging signs, early and late outcomes, and response

to intravenous alteplase after acute ischaemic stroke in the third International Stroke

Trial (IST-3): secondary analysis of a randomised controlled trial. The Lancet

Neurology, 2015, 14(5): 485-496.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 230

[350] CHARIDIMOU A, PASI M, FIORELLI M, et al. Leukoaraiosis, Cerebral

Haemorrhage, and Outcome After Intravenous Thrombolysis for Acute Ischemic

Stroke: A Meta-Analysis (v1). Stroke; a journal of cerebral circulation, 2016, 47(9):

2364-2372.

[351] MAIR G, VON KUMMER R, MORRIS Z, et al. Effect of alteplase on the CT

hyperdense artery sign and outcome after ischemic stroke. Neurology, 2016, 86(2):

118-125.

[352] CHARIDIMOU A, SHOAMANESH A. Clinical relevance of microbleeds in

acute stroke thrombolysis: Comprehensive meta-analysis. Neurology, 2016, 87(15):

1534-1541.

[353] SMITH E E, KENT D M, BULSARA K R, et al. Accuracy of Prediction

Instruments for Diagnosing Large Vessel Occlusion in Individuals With Suspected

Stroke: A Systematic Review for the 2018 Guidelines for the Early Management of

Patients With Acute Ischemic Stroke. Stroke; a journal of cerebral circulation, 2018,

49(3): e111-e122.

[354] EHRLICH M E, TURNER H L, CURRIE L J, et al. Safety of Computed

Tomographic Angiography in the Evaluation of Patients With Acute Stroke: A Single-

Center Experience. Stroke; a journal of cerebral circulation, 2016, 47(8): 2045-2050.

[355] LIMA F O, LEV M H, LEVY R A, et al. Functional contrast-enhanced CT for

evaluation of acute ischemic stroke does not increase the risk of contrast-induced

nephropathy. AJNR American journal of neuroradiology, 2010, 31(5): 817-821.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 231

[356] AULICKY P, MIKULIK R, GOLDEMUND D, et al. Safety of performing CT

angiography in stroke patients treated with intravenous thrombolysis. Journal of

neurology, neurosurgery, and psychiatry, 2010, 81(7): 783-787.

[357] WITT B J, BALLMAN K V, BROWN R D, JR., et al. The incidence of stroke

after myocardial infarction: a meta-analysis. Am J Med, 2006, 119(4): 354 e351-359.

[358] OSHEROV A B, BOROVIK-RAZ M, ARONSON D, et al. Incidence of early

left ventricular thrombus after acute anterior wall myocardial infarction in the primary

coronary intervention era. American heart journal, 2009, 157(6): 1074-1080.

[359] SOLHEIM S, SELJEFLOT I, LUNDE K, et al. Frequency of left ventricular

thrombus in patients with anterior wall acute myocardial infarction treated with

percutaneous coronary intervention and dual antiplatelet therapy. The American

journal of cardiology, 2010, 106(9): 1197-1200.

[360] SCHWALM J D, AHMAD M, SALEHIAN O, et al. Warfarin after anterior

myocardial infarction in current era of dual antiplatelet therapy: a randomized

feasibility trial. J Thromb Thrombolysis, 2010, 30(2): 127-132.

[361] VAITKUS P T, BARNATHAN E S. Embolic potential, prevention and

management of mural thrombus complicating anterior myocardial infarction: a meta-

analysis. J Am Coll Cardiol, 1993, 22(4): 1004-1009.

[362] CLELAND J G, FINDLAY I, JAFRI S, et al. The Warfarin/Aspirin Study in

Heart failure (WASH): a randomized trial comparing antithrombotic strategies for

patients with heart failure. American heart journal, 2004, 148(1): 157-164.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 232

[363] MASSIE B M, COLLINS J F, AMMON S E, et al. Randomized trial of

warfarin, aspirin, and clopidogrel in patients with chronic heart failure: the Warfarin

and Antiplatelet Therapy in Chronic Heart Failure (WATCH) trial. Circulation, 2009,

119(12): 1616-1624.

[364] COKKINOS D V, HARALABOPOULOS G C, KOSTIS J B, et al. Efficacy of

antithrombotic therapy in chronic heart failure: the HELAS study. Eur J Heart Fail,

2006, 8(4): 428-432.

[365] MORAN J F. Neurologic complications of cardiomyopathies and other

myocardial disorders. Handb Clin Neurol, 2014, 119(111-128.

[366] WHITLOCK R P, SUN J C, FREMES S E, et al. Antithrombotic and

thrombolytic therapy for valvular disease: Antithrombotic Therapy and Prevention of

Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical

Practice Guidelines. Chest, 2012, 141(2 Suppl): e576S-e600S.

[367] BARLETTA G A, GAGLIARDI R, BENVENUTI L, et al. Cerebral ischemic

attacks as a complication of aortic and mitral valve prolapse. Stroke, 1985, 16(2):

219-223.

[368] GILON D, BUONANNO F S, JOFFE M M, et al. Lack of evidence of an

association between mitral-valve prolapse and stroke in young patients. N Engl J

Med, 1999, 341(1): 8-13.

[369] FREED L A, LEVY D, LEVINE R A, et al. Prevalence and clinical outcome of

mitral-valve prolapse. N Engl J Med, 1999, 341(1): 1-7.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 233

[370] AVIERINOS J F, BROWN R D, FOLEY D A, et al. Cerebral ischemic events

after diagnosis of mitral valve prolapse: a community-based study of incidence and

predictive factors. Stroke, 2003, 34(6): 1339-1344.

[371] BENJAMIN E J, PLEHN J F, D'AGOSTINO R B, et al. Mitral annular

calcification and the risk of stroke in an elderly cohort. N Engl J Med, 1992, 327(6):

374-379.

[372] KIZER J R, WIEBERS D O, WHISNANT J P, et al. Mitral annular

calcification, aortic valve sclerosis, and incident stroke in adults free of clinical

cardiovascular disease: the Strong Heart Study. Stroke, 2005, 36(12): 2533-2537.

[373] KOHSAKA S, JIN Z, RUNDEK T, et al. Impact of mitral annular calcification

on cardiovascular events in a multiethnic community: the Northern Manhattan Study.

JACC Cardiovasc Imaging, 2008, 1(5): 617-623.

[374] JILAIHAWI H, CHAKRAVARTY T, WEISS R E, et al. Meta-analysis of

complications in aortic valve replacement: comparison of Medtronic-Corevalve,

Edwards-Sapien and surgical aortic valve replacement in 8,536 patients. Catheter

Cardiovasc Interv, 2012, 80(1): 128-138.

[375] BONOW R O, CARABELLO B A, CHATTERJEE K, et al. 2008 Focused

update incorporated into the ACC/AHA 2006 guidelines for the management of

patients with valvular heart disease: a report of the American College of

Cardiology/American Heart Association Task Force on Practice Guidelines (Writing

Committee to Revise the 1998 Guidelines for the Management of Patients With

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 234

Valvular Heart Disease): endorsed by the Society of Cardiovascular Anesthesiologists,

Society for Cardiovascular Angiography and Interventions, and Society of Thoracic

Surgeons. Circulation, 2008, 118(15): e523-661.

[376] HERAS M, CHESEBRO J H, FUSTER V, et al. High risk of thromboemboli

early after bioprosthetic cardiac valve replacement. J Am Coll Cardiol, 1995, 25(5):

1111-1119.

[377] MYLONAKIS E, CALDERWOOD S B. Infective endocarditis in adults. N

Engl J Med, 2001, 345(18): 1318-1330.

[378] THUNY F, DI SALVO G, BELLIARD O, et al. Risk of embolism and death in

infective endocarditis: prognostic value of echocardiography: a prospective

multicenter study. Circulation, 2005, 112(1): 69-75.

[379] VILACOSTA I, GRAUPNER C, SAN ROMAN J A, et al. Risk of embolization

after institution of antibiotic therapy for infective endocarditis. J Am Coll Cardiol,

2002, 39(9): 1489-1495.

[380] DICKERMAN S A, ABRUTYN E, BARSIC B, et al. The relationship between

the initiation of antimicrobial therapy and the incidence of stroke in infective

endocarditis: an analysis from the ICE Prospective Cohort Study (ICE-PCS).

American heart journal, 2007, 154(6): 1086-1094.

[381] HEIRO M, NIKOSKELAINEN J, ENGBLOM E, et al. Neurologic

manifestations of infective endocarditis: a 17-year experience in a teaching hospital in

Finland. Archives of internal medicine, 2000, 160(18): 2781-2787.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 235

[382] CABELL C H, POND K K, PETERSON G E, et al. The risk of stroke and death

in patients with aortic and mitral valve endocarditis. American heart journal, 2001,

142(1): 75-80.

[383] ACEBO E, VAL-BERNAL J F, GOMEZ-ROMAN J J, et al. Clinicopathologic

study and DNA analysis of 37 cardiac myxomas: a 28-year experience. Chest, 2003,

123(5): 1379-1385.

[384] GOWDA R M, KHAN I A, NAIR C K, et al. Cardiac papillary fibroelastoma: a

comprehensive analysis of 725 cases. American heart journal, 2003, 146(3): 404-410.

[385] ROBERTS W C. Primary and secondary neoplasms of the heart. The American

journal of cardiology, 1997, 80(5): 671-682.

[386] REYNEN K. Cardiac myxomas. N Engl J Med, 1995, 333(24): 1610-1617.

[387] HOWARD R A, ALDEA G S, SHAPIRA O M, et al. Papillary fibroelastoma:

increasing recognition of a surgical disease. Ann Thorac Surg, 1999, 68(5): 1881-

1885.

[388] EKMEKTZOGLOU K A, SAMELIS G F, XANTHOS T. Heart and tumors:

location, metastasis, clinical manifestations, diagnostic approaches and therapeutic

considerations. J Cardiovasc Med (Hagerstown), 2008, 9(8): 769-777.

[389] AMARENCO P, COHEN A, TZOURIO C, et al. Atherosclerotic disease of the

aortic arch and the risk of ischemic stroke. N Engl J Med, 1994, 331(22): 1474-1479.

[390] DI TULLIO M R, RUSSO C, JIN Z, et al. Aortic arch plaques and risk of

recurrent stroke and death. Circulation, 2009, 119(17): 2376-2382.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 236

[391] MACKENSEN G B, TI L K, PHILLIPS-BUTE B G, et al. Cerebral

embolization during cardiac surgery: impact of aortic atheroma burden. Br J Anaesth,

2003, 91(5): 656-661.

[392] KRONZON I, TUNICK P A. Aortic atherosclerotic disease and stroke.

Circulation, 2006, 114(1): 63-75.

[393] FRENCH STUDY OF AORTIC PLAQUES IN STROKE G, AMARENCO P,

COHEN A, et al. Atherosclerotic disease of the aortic arch as a risk factor for

recurrent ischemic stroke. N Engl J Med, 1996, 334(19): 1216-1221.

[394] DOTY J R, WILENTZ R E, SALAZAR J D, et al. Atheroembolism in cardiac

surgery. Ann Thorac Surg, 2003, 75(4): 1221-1226.

[395] BUTCHART E G, MORENO DE LA SANTA P, ROONEY S J, et al. The role

of risk factors and trigger factors in cerebrovascular events after mitral valve

replacement: implications for antithrombotic management. J Card Surg, 1994, 9(2

Suppl): 228-236.

[396] AHLSSON A, FENGSRUD E, BODIN L, et al. Postoperative atrial fibrillation

in patients undergoing aortocoronary bypass surgery carries an eightfold risk of future

atrial fibrillation and a doubled cardiovascular mortality. Eur J Cardiothorac Surg,

2010, 37(6): 1353-1359.

[397] BUBER J, LURIA D, STERNIK L, et al. Morphological features of the P-

waves at surface electrocardiogram as surrogate to mechanical function of the left

atrium following a successful modified maze procedure. Europace, 2014, 16(4): 578-

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 237

586.

[398] BANDO K, KOBAYASHI J, HIRATA M, et al. Early and late stroke after

mitral valve replacement with a mechanical prosthesis: risk factor analysis of a 24-

year experience. J Thorac Cardiovasc Surg, 2003, 126(2): 358-364.

[399] COX J L, AD N, PALAZZO T. Impact of the maze procedure on the stroke rate

in patients with atrial fibrillation. J Thorac Cardiovasc Surg, 1999, 118(5): 833-840.

[400] MIN X P, ZHU T Y, HAN J, et al. Left atrial appendage obliteration in atrial

fibrillation patients undergoing bioprosthetic mitral valve replacement. Herz, 2016,

41(1): 87-94.

[401] TOMSON T T, KAPA S, BALA R, et al. Risk of stroke and atrial fibrillation

after radiofrequency catheter ablation of typical atrial flutter. Heart Rhythm, 2012,

9(11): 1779-1784.

[402] BUTCHART E G, PAYNE N, LI H H, et al. Better anticoagulation control

improves survival after valve replacement. J Thorac Cardiovasc Surg, 2002, 123(4):

715-723.

[403] STRANDBERG M, MARTTILA R J, HELENIUS H, et al. Transoesophageal

echocardiography in selecting patients for anticoagulation after ischaemic stroke or

transient ischaemic attack. Journal of neurology, neurosurgery, and psychiatry, 2002,

73(1): 29-33.

[404] YAMAMOTO M, SEO Y, KAWAMATSU N, et al. Complex left atrial

appendage morphology and left atrial appendage thrombus formation in patients with

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 238

atrial fibrillation. Circ Cardiovasc Imaging, 2014, 7(2): 337-343.

[405] BAHER A, MOWLA A, KODALI S, et al. Cardiac MRI improves

identification of aetiology of acute ischemic stroke. Cerebrovasc Dis, 2014, 37(4):

277-284.

[406] WEINSAFT J W, KIM H W, CROWLEY A L, et al. LV thrombus detection by

routine echocardiography: insights into performance characteristics using delayed

enhancement CMR. JACC Cardiovasc Imaging, 2011, 4(7): 702-712.

[407] WALTERS T E, ELLIMS A H, KALMAN J M. The role of left atrial imaging in

the management of atrial fibrillation. Prog Cardiovasc Dis, 2015, 58(2): 136-151.

[408] AKOUM N, FERNANDEZ G, WILSON B, et al. Association of atrial fibrosis

quantified using LGE-MRI with atrial appendage thrombus and spontaneous contrast

on transesophageal echocardiography in patients with atrial fibrillation. J Cardiovasc

Electrophysiol, 2013, 24(10): 1104-1109.

[409] FONSECA A C, ALVES P, INACIO N, et al. Patients With Undetermined

Stroke Have Increased Atrial Fibrosis: A Cardiac Magnetic Resonance Imaging Study.

Stroke, 2018, 49(3): 734-737.

[410] TSE H F, WANG Y J, AHMED AI-ABDULLAH M, et al. Stroke prevention in

atrial fibrillation--an Asian stroke perspective. Heart Rhythm, 2013, 10(7): 1082-

1088.

[411] WONG C X, BROWN A, TSE H F, et al. Epidemiology of Atrial Fibrillation:

The Australian and Asia-Pacific Perspective. Heart Lung Circ, 2017, 26(9): 870-879.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 239

[412] KANNEL W B, BENJAMIN E J. Status of the epidemiology of atrial

fibrillation. Med Clin North Am, 2008, 92(1): 17-40, ix.

[413] KWAN J, SANDERCOCK P. In-hospital care pathways for stroke: a Cochrane

systematic review. Stroke, 2003, 34(2): 587-588.

[414] SUZUKI M, IMAI A, HONDA M, et al. Role of a critical pathway for door-to-

CT-completion interval in the management of acute ischemic stroke patients in the

emergency room. Keio J Med, 2004, 53(4): 247-250.

[415] MEHDIRATTA M, WOOLFENDEN A R, CHAPMAN K M, et al. Reduction in

IV t-PA door to needle times using an Acute Stroke Triage Pathway. Can J Neurol Sci,

2006, 33(2): 214-216.

[416] PATEL P J, KATZ R, BOROVSKIY Y, et al. Race and stroke in an atrial

fibrillation inception cohort: Findings from the Penn Atrial Fibrillation Free study.

Heart Rhythm, 2018, 15(4): 487-493.

[417] BRITTON M, GUSTAFSSON C. Non-rheumatic atrial fibrillation as a risk

factor for stroke. Stroke, 1985, 16(2): 182-188.

[418] APPELROS P, NYDEVIK I, VIITANEN M. Poor outcome after first-ever

stroke: predictors for death, dependency, and recurrent stroke within the first year.

Stroke, 2003, 34(1): 122-126.

[419] LIN H J, WOLF P A, KELLY-HAYES M, et al. Stroke severity in atrial

fibrillation. The Framingham Study. Stroke, 1996, 27(10): 1760-1764.

[420] MIYASAKA Y, BARNES M E, GERSH B J, et al. Time trends of ischemic

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 240

stroke incidence and mortality in patients diagnosed with first atrial fibrillation in

1980 to 2000: report of a community-based study. Stroke, 2005, 36(11): 2362-2366.

[421] SCHWAMMENTHAL Y, BORNSTEIN N, SCHWAMMENTHAL E, et al.

Relation of effective anticoagulation in patients with atrial fibrillation to stroke

severity and survival (from the National Acute Stroke Israeli Survey [NASIS]). The

American journal of cardiology, 2010, 105(3): 411-416.

[422] WANG Y, MINEMATSU K, WONG K S, et al. Ticagrelor in Acute Stroke or

Transient Ischemic Attack in Asian Patients: From the SOCRATES Trial (Acute

Stroke or Transient Ischemic Attack Treated With Aspirin or Ticagrelor and Patient

Outcomes). Stroke, 2017, 48(1): 167-173.

[423] NACCARELLI G V, JOHNSTON S S, LIN J, et al. Cost burden of

cardiovascular hospitalization and mortality in ATHENA-like patients with atrial

fibrillation/atrial flutter in the United States. Clin Cardiol, 2010, 33(5): 270-279.

[424] LUO J, LI H, QIN X, et al. Increased risk of ischemic stroke associated with

new-onset atrial fibrillation complicating acute coronary syndrome: A systematic

review and meta-analysis. Int J Cardiol, 2018,

[425] BURUP KRISTENSEN C, JENSEN J S, SOGAARD P, et al. Atrial fibrillation

in aortic stenosis--echocardiographic assessment and prognostic importance.

Cardiovasc Ultrasound, 2012, 10(38.

[426] JOHNSON L S B, PERSSON A P, WOLLMER P, et al. Irregularity and lack of

p waves in short tachycardia episodes predict atrial fibrillation and ischemic stroke.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 241

Heart Rhythm, 2018,

[427] NIEUWLAAT R, PRINS M H, LE HEUZEY J Y, et al. Prognosis, disease

progression, and treatment of atrial fibrillation patients during 1 year: follow-up of the

Euro Heart Survey on atrial fibrillation. Eur Heart J, 2008, 29(9): 1181-1189.

[428] MANT J, HOBBS F D, FLETCHER K, et al. Warfarin versus aspirin for stroke

prevention in an elderly community population with atrial fibrillation (the

Birmingham Atrial Fibrillation Treatment of the Aged Study, BAFTA): a randomised

controlled trial. Lancet, 2007, 370(9586): 493-503.

[429] WOLF P A, ABBOTT R D, KANNEL W B. Atrial fibrillation as an

independent risk factor for stroke: the Framingham Study. Stroke, 1991, 22(8): 983-

988.

[430] WOLF P A, MITCHELL J B, BAKER C S, et al. Impact of atrial fibrillation on

mortality, stroke, and medical costs. Archives of internal medicine, 1998, 158(3): 229-

234.

[431] WANG C, LIU Y, YANG Q, et al. Body mass index and risk of total and type-

specific stroke in Chinese adults: results from a longitudinal study in China.

International journal of stroke : official journal of the International Stroke Society,

2013, 8(4): 245-250.

[432] KAMEL H, ELKIND M S, BHAVE P D, et al. Paroxysmal supraventricular

tachycardia and the risk of ischemic stroke. Stroke, 2013, 44(6): 1550-1554.

[433] ARBOIX A, MARTI L, DORISON S, et al. Bayes syndrome and acute

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 242

cardioembolic ischemic stroke. World journal of clinical cases, 2017, 5(3): 93-101.

[434] ARIYARAJAH V, PURI P, APIYASAWAT S, et al. Interatrial block: a novel

risk factor for embolic stroke? Annals of noninvasive electrocardiology : the official

journal of the International Society for Holter and Noninvasive Electrocardiology,

Inc, 2007, 12(1): 15-20.

[435] CHHABRA L, SRINIVASAN I, SAREEN P, et al. Interatrial block - a novel

risk factor for acute mesenteric ischemia. Indian J Gastroenterol, 2012, 31(4): 191-

194.

[436] LORBAR M, LEVRAULT R, PHADKE J G, et al. Interatrial block as a

predictor of embolic stroke. The American journal of cardiology, 2005, 95(5): 667-

668.

[437] LIP G Y. Implications of the CHA(2)DS(2)-VASc and HAS-BLED Scores for

thromboprophylaxis in atrial fibrillation. Am J Med, 2011, 124(2): 111-114.

[438] STROKE RISK IN ATRIAL FIBRILLATION WORKING G. Comparison of

12 risk stratification schemes to predict stroke in patients with nonvalvular atrial

fibrillation. Stroke, 2008, 39(6): 1901-1910.

[439] WALDO A L. Anticoagulation: stroke prevention in patients with atrial

fibrillation. Cardiol Clin, 2009, 27(1): 125-135, ix.

[440] GAGE B F, WATERMAN A D, SHANNON W, et al. Validation of clinical

classification schemes for predicting stroke: results from the National Registry of

Atrial Fibrillation. Jama, 2001, 285(22): 2864-2870.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 243

[441] LIP G Y, HALPERIN J L. Improving stroke risk stratification in atrial

fibrillation. Am J Med, 2010, 123(6): 484-488.

[442] BORIANI G, BOTTO G L, PADELETTI L, et al. Improving stroke risk

stratification using the CHADS2 and CHA2DS2-VASc risk scores in patients with

paroxysmal atrial fibrillation by continuous arrhythmia burden monitoring. Stroke,

2011, 42(6): 1768-1770.

[443] TAILLANDIER S, OLESEN J B, CLEMENTY N, et al. Prognosis in patients

with atrial fibrillation and CHA2DS2-VASc Score = 0 in a community-based cohort

study. J Cardiovasc Electrophysiol, 2012, 23(7): 708-713.

[444] CHAO T F, LIU C J, WANG K L, et al. Incidence and prediction of ischemic

stroke among atrial fibrillation patients with end-stage renal disease requiring dialysis.

Heart Rhythm, 2014, 11(10): 1752-1759.

[445] WOLSK E, LAMBERTS M, HANSEN M L, et al. Thromboembolic risk

stratification of patients hospitalized with heart failure in sinus rhythm: a nationwide

cohort study. Eur J Heart Fail, 2015, 17(8): 828-836.

[446] FITZMAURICE D A, HOBBS F D, JOWETT S, et al. Screening versus routine

practice in detection of atrial fibrillation in patients aged 65 or over: cluster

randomised controlled trial. BMJ, 2007, 335(7616): 383.

[447] HOBBS F D, FITZMAURICE D A, MANT J, et al. A randomised controlled

trial and cost-effectiveness study of systematic screening (targeted and total

population screening) versus routine practice for the detection of atrial fibrillation in

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 244

people aged 65 and over. The SAFE study. Health Technol Assess, 2005, 9(40): iii-iv,

ix-x, 1-74.

[448] RABINSTEIN A A. Prolonged cardiac monitoring for detection of paroxysmal

atrial fibrillation after cerebral ischemia. Stroke, 2014, 45(4): 1208-1214.

[449] WIESEL J, FITZIG L, HERSCHMAN Y, et al. Detection of atrial fibrillation

using a modified microlife blood pressure monitor. Am J Hypertens, 2009, 22(8): 848-

852.

[450] WACHTER R, GROSCHEL K, GELBRICH G, et al. Holter-electrocardiogram-

monitoring in patients with acute ischaemic stroke (Find-AFRANDOMISED): an

open-label randomised controlled trial. The Lancet Neurology, 2017, 16(4): 282-290.

[451] GLADSTONE D J, SPRING M, DORIAN P, et al. Atrial fibrillation in patients

with cryptogenic stroke. The New England journal of medicine, 2014, 370(26): 2467-

2477.

[452] SANNA T, DIENER H C, PASSMAN R S, et al. Cryptogenic stroke and

underlying atrial fibrillation. N Engl J Med, 2014, 370(26): 2478-2486.

[453] GLOTZER T V, DAOUD E G, WYSE D G, et al. The relationship between

daily atrial tachyarrhythmia burden from implantable device diagnostics and stroke

risk: the TRENDS study. Circulation Arrhythmia and electrophysiology, 2009, 2(5):

474-480.

[454] STEINHUBL S R, MEHTA R R, EBNER G S, et al. Rationale and design of a

home-based trial using wearable sensors to detect asymptomatic atrial fibrillation in a

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 245

targeted population: The mHealth Screening To Prevent Strokes (mSToPS) trial.

American heart journal, 2016, 175(77-85.

[455] MUSE E D, WINEINGER N E, SPENCER E G, et al. Validation of a genetic

risk score for atrial fibrillation: A prospective multicenter cohort study. PLoS Med,

2018, 15(3): e1002525.

[456] FUKUSHIMA K, FUKUSHIMA N, KATO K, et al. Correlation between left

atrial appendage morphology and flow velocity in patients with paroxysmal atrial

fibrillation. Eur Heart J Cardiovasc Imaging, 2016, 17(1): 59-66.

[457] GOLDMAN M E, PEARCE L A, HART R G, et al. Pathophysiologic correlates

of thromboembolism in nonvalvular atrial fibrillation: I. Reduced flow velocity in the

left atrial appendage (The Stroke Prevention in Atrial Fibrillation [SPAF-III] study). J

Am Soc Echocardiogr, 1999, 12(12): 1080-1087.

[458] RIES D L, ROSENBERG Y D, WACLAWIW M A, et al. Ejection fraction and

risk of thromboembolic events in patients with systolic dysfunction and sinus rhythm:

evidence for gender differences in the studies of left ventricular dysfunction trials. J

Am Coll Cardiol, 1997, 29(5): 1074-1080.

[459] GUPTA D K, SHAH A M, GIUGLIANO R P, et al. Left atrial structure and

function in atrial fibrillation: ENGAGE AF-TIMI 48. Eur Heart J, 2014, 35(22):

1457-1465.

[460] BELEN E, OZAL E, PUSUROGLU H. Association of the CHA2DS2-VASc

score with left atrial spontaneous echo contrast: a cross-sectional study of patients

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 246

with rheumatic mitral stenosis in sinus rhythm. Heart Vessels, 2016, 31(9): 1537-

1543.

[461] HANDKE M, HARLOFF A, HETZEL A, et al. Predictors of left atrial

spontaneous echocardiographic contrast or thrombus formation in stroke patients with

sinus rhythm and reduced left ventricular function. The American journal of

cardiology, 2005, 96(9): 1342-1344.

[462] AMMASH N, KONIK E A, MCBANE R D, et al. Left atrial blood stasis and

Von Willebrand factor-ADAMTS13 homeostasis in atrial fibrillation. Arterioscler

Thromb Vasc Biol, 2011, 31(11): 2760-2766.

[463] SCRIDON A, GIRERD N, RUGERI L, et al. Progressive endothelial damage

revealed by multilevel von Willebrand factor plasma concentrations in atrial

fibrillation patients. Europace, 2013, 15(11): 1562-1566.

[464] CHUNG H, JOUNG B, LEE K Y, et al. Left Atrial Volume Index Predicts

Recurrence of Stroke in Patients with Nonsustained Atrial Tachycardia. Journal of

stroke and cerebrovascular diseases : the official journal of National Stroke

Association, 2015, 24(10): 2408-2415.

[465] HART R G, DIENER H C, COUTTS S B, et al. Embolic strokes of

undetermined source: the case for a new clinical construct. The Lancet Neurology,

2014, 13(4): 429-438.

[466] YONG M, DIENER H C, KASTE M, et al. Characteristics of blood pressure

profiles as predictors of long-term outcome after acute ischemic stroke. Stroke, 2005,

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 247

36(12): 2619-2625.

[467] STEAD L G, GILMORE R M, VEDULA K C, et al. Impact of acute blood

pressure variability on ischemic stroke outcome. Neurology, 2006, 66(12): 1878-

1881.

[468] MONIES D, ABOUELHODA M, ALSAYED M, et al. The landscape of genetic

diseases in Saudi Arabia based on the first 1000 diagnostic panels and exomes.

Human genetics, 2017, 136(8): 921-939.

[469] KAKALETSIS N, NTAIOS G, MILIONIS H, et al. Prognostic value of 24-h

ABPM in acute ischemic stroke for short-, medium-, and long-term outcome: a

systematic review and meta-analysis. International journal of stroke : official journal

of the International Stroke Society, 2015, 10(7): 1000-1007.

[470] CHUNG J W, KIM N, KANG J, et al. Blood pressure variability and the

development of early neurological deterioration following acute ischemic stroke.

Journal of hypertension, 2015, 33(10): 2099-2106.

[471] FAN Z X, YE X G, HUANG J L.Correlation between changes in blood pressure

and neurological deterioration in the early post-acute ischemic stroke. Chinese

Journal of Modern Drug Application, 2017, 8): 82-83.

[472] SUN Y M. The Relationship between Blood Pressure Changes and Early

Neurological Deterioration in Patients with Acute Ischemic Stroke. Journal of Shanxi

Datong University(Natural Science Edition), 2017, 06): 47-49.

[473] CASTILLO J, LEIRA R, GARCIA M M, et al. Blood pressure decrease during

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 248

the acute phase of ischemic stroke is associated with brain injury and poor stroke

outcome. Stroke, 2004, 35(2): 520-526.

[474] TIEN Y T, CHANG M H, LEE Y S, et al. Pulse Blood Pressure Correlates with

Late Outcome in Acute Ischemic Stroke without Significant Culprit Artery Stenosis.

Journal of stroke and cerebrovascular diseases : the official journal of National Stroke

Association, 2016, 25(5): 1229-1234.

[475] LEE K J, KIM B J, HAN M K, et al. Predictive Value of Pulse Pressure in

Acute Ischemic Stroke for Future Major Vascular Events. Stroke, 2018, 49(1): 46-53.

[476] JUSUFOVIC M, SANDSET E C, BATH P M, et al. Effects of blood pressure

lowering in patients with acute ischemic stroke and carotid artery stenosis.

International journal of stroke : official journal of the International Stroke Society,

2015, 10(3): 354-359.

[477] AMARENCO P, BOGOUSSLAVSKY J, CALLAHAN A, 3RD, et al. High-

dose atorvastatin after stroke or transient ischemic attack. N Engl J Med, 2006,

355(6): 549-559.

[478] WANG M X, CHEN G, ZHOU W S, et al. Relationship between lipids and

ABCD2 score in patients with transient ischemic attack. Progress in Modern

Biomedicine, 2011, 11): 2138-2140.

[479] WANG G Z, ZHANG C, WANG G H, et al. Correlation of acute ischemic

stroke with cerebral hemorrhage and glycated hemoglobin and lipids. Chinese Journal

of Clinical Neurosciences, 2013, 04): 438-442.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 249

[480] SONG P, ZHANG Q S, SUN H Q. Comparison of clinical efficacy of different

doses of atorvastatin in the treatment of acute ischemic stroke. Journal of Practical

Cardiocerebral Pulmonary Vasculopathy, 2016, 10): 155-157.

[481] WU J L. Multivariate analysis of enhanced lipid-lowering for patients with

TOAST classification of acute ischemic stroke [D]; Nanchang University, 2016.

[482] GOLDSTEIN L B, AMARENCO P, SZAREK M, et al. Haemorrhagic stroke in

the Stroke Prevention by Aggressive Reduction in Cholesterol Levels study.

Neurology, 2008, 70(24 Pt 2): 2364-2370.

[483] ZHOU M, PANG G F, JIANG A L, et al. Study on the relationship between

lipid metabolism and cerebral hemorrhage transformation in the patients with

ischemic stroke. China Modern Doctor, 2016, 28): 1-4,8.

[484] TANG Y Y, QIN C, LIANG Z J, et al. Study on the relationship of hemorrhagic

transformation after acute ischemic stroke and the level of fasting blood lipid. Chinese

Journal of Nervous and Mental Diseases, 2011, 12): 740-743.

[485] LI Z X, ZHANG Q Q, LI C. Correlative analysis of glycosylated hemoglobin ,

blood lipid and acute ischemic stroke with cerebral microbleeds in elderly patients.

Journal of Clinical Psychosomatic Diseases, 2018, 3): 8-11.

[486] LI W, LIU M, WANG L C, et al. Relation between the serum lipid level and

state of severity in patients with stroke in early stage. Journal of Clinical Neurology,

2007, 3): 222-223.

[487] CHEN F. Relationship between blood lipid level and neurologic impairment in

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 250

acute stage of cerebral stroke. Practical Journal of Clinical Medicine, 2008, 5): 114-

115.

[488] HUANG P. Correlation between blood lipid levels and prognosis of acute

ischemic stroke [D]; Suzhou University, 2016.

[489] GU S E. Study on the relationship between blood lipid levels and prognosis in

the acute phase of ischemic stroke [D]; Ningxia Medical University, 2011.

[490] YANG J, ABDUSALAM A, ZHANG L. Study on the association of serum ox-

LDL and Lp-PLA2 levels with atherosclerosis and neurological deficits in patients

with cerebral ischemic stroke. International Journal of Laboratory Medicine, 2017,

23): 3283-3285.

[491] GU S E, YANG P, MA D W, et al. Correlation between blood lipid levels and

prognosis in patients with acute ischemic stroke. Journal of Ningxia Medical College,

2011, 5): 470-472.

[492] WANG C J, WANG Y L, LI Z X, et al. The Management of LDL Cholesterol

and Predictors of Goal Achievement in Stroke Patients in China: A Cross-Sectional

Study. CNS neuroscience & therapeutics, 2016, 22(7): 577-583.

[493] SMITH E E, ABDULLAH A R, AMIRFARZAN H, et al. Serum lipid profile on

admission for ischemic stroke: failure to meet National Cholesterol Education

Program Adult Treatment Panel (NCEP-ATPIII) guidelines. Neurology, 2007, 68(9):

660-665.

[494] SAPOSNIK G, FONAROW G C, PAN W, et al. Guideline-directed low-density

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 251

lipoprotein management in high-risk patients with ischemic stroke: findings from Get

with the Guidelines-Stroke 2003 to 2012. Stroke, 2014, 45(11): 3343-3351.

[495] MCALISTER F A, MAJUMDAR S R, PADWAL R S, et al. Case management

for blood pressure and lipid level control after minor stroke: PREVENTION

randomized controlled trial. CMAJ : Canadian Medical Association journal = journal

de l'Association medicale canadienne, 2014, 186(8): 577-584.

[496] OSEI E, DEN HERTOG H M, BERKHEMER O A, et al. Increased admission

and fasting glucose are associated with unfavorable short-term outcome after intra-

arterial treatment of ischemic stroke in the MR CLEAN pretrial cohort. J Neurol Sci,

2016, 371(1-5.

[497] CRUZ-HERRANZ A, FUENTES B, MARTINEZ-SANCHEZ P, et al. Is

diabetes an independent risk factor for in-hospital complications after a stroke?

Journal of diabetes, 2015, 7(5): 657-663.

[498] PAN Y, CHEN W, JING J, et al. Pancreatic beta-Cell Function and Prognosis of

Nondiabetic Patients With Ischemic Stroke. Stroke, 2017, 48(11): 2999-3005.

[499] WALTERS M R, WEIR C J, LEES K R. A randomised, controlled pilot study to

investigate the potential benefit of intervention with insulin in hyperglycaemic acute

ischaemic stroke patients. Cerebrovasc Dis, 2006, 22(2-3): 116-122.

[500] STASZEWSKI J, BRODACKI B, KOTOWICZ J, et al. Intravenous insulin

therapy in the maintenance of strict glycemic control in nondiabetic acute stroke

patients with mild hyperglycemia. Journal of stroke and cerebrovascular diseases : the

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 252

official journal of National Stroke Association, 2011, 20(2): 150-154.

[501] MCCORMICK M, HADLEY D, MCLEAN J R, et al. Randomized, controlled

trial of insulin for acute poststroke hyperglycemia. Annals of neurology, 2010, 67(5):

570-578.

[502] KERNAN W N, VISCOLI C M, BRASS L M, et al. The stroke prognosis

instrument II (SPI-II) : A clinical prediction instrument for patients with transient

ischemia and nondisabling ischemic stroke. Stroke, 2000, 31(2): 456-462.

[503] WEIMAR C, DIENER H C, ALBERTS M J, et al. The Essen stroke risk score

predicts recurrent cardiovascular events: a validation within the REduction of

Atherothrombosis for Continued Health (REACH) registry. Stroke, 2009, 40(2): 350-

354.

[504] TSIVGOULIS G, VASSILOPOULOU S, SPENGOS K. Potential applicability

of ABCD score in triaging TIA patients. Lancet, 2007, 369(9567): 1082.

[505] WARDLAW J M, BRAZZELLI M, CHAPPELL F M, et al. ABCD2 score and

secondary stroke prevention: meta-analysis and effect per 1,000 patients triaged.

Neurology, 2015, 85(4): 373-380.

[506] KELLY P J, ALBERS G W, CHATZIKONSTANTINOU A, et al. Validation and

comparison of imaging-based scores for prediction of early stroke risk after transient

ischaemic attack: a pooled analysis of individual-patient data from cohort studies. The

Lancet Neurology, 2016, 15(12): 1238-1247.

[507] KIYOHARA T, KAMOUCHI M, KUMAI Y, et al. ABCD3 and ABCD3-I

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 253

scores are superior to ABCD2 score in the prediction of short- and long-term risks of

stroke after transient ischemic attack. Stroke, 2014, 45(2): 418-425.

[508] LIP G Y, FRISON L, HALPERIN J L, et al. Identifying patients at high risk for

stroke despite anticoagulation: a comparison of contemporary stroke risk stratification

schemes in an anticoagulated atrial fibrillation cohort. Stroke, 2010, 41(12): 2731-

2738.

[509] LIP G Y, NIEUWLAAT R, PISTERS R, et al. Refining clinical risk

stratification for predicting stroke and thromboembolism in atrial fibrillation using a

novel risk factor-based approach: the euro heart survey on atrial fibrillation. Chest,

2010, 137(2): 263-272.

[510] RIETBROCK S, HEELEY E, PLUMB J, et al. Chronic atrial fibrillation:

Incidence, prevalence, and prediction of stroke using the Congestive heart failure,

Hypertension, Age >75, Diabetes mellitus, and prior Stroke or transient ischemic

attack (CHADS2) risk stratification scheme. American heart journal, 2008, 156(1):

57-64.

[511] PISTERS R, LANE D A, NIEUWLAAT R, et al. A novel user-friendly score

(HAS-BLED) to assess 1-year risk of major bleeding in patients with atrial

fibrillation: the Euro Heart Survey. Chest, 2010, 138(5): 1093-1100.

[512] ANDERSSON T, KUNTZE SODERQVIST A, SODERMAN M, et al.

Mechanical thrombectomy as the primary treatment for acute basilar artery occlusion:

experience from 5 years of practice. J Neurointerv Surg, 2013, 5(3): 221-225.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 254

[513] GAO S, WANG Y J, XU A D, et al. Chinese ischemic stroke subclassification.

Frontiers in neurology, 2011, 2(6.

[514] CHIMOWITZ M I, LYNN M J, DERDEYN C P, et al. Stenting versus

aggressive medical therapy for intracranial arterial stenosis. N Engl J Med, 2011,

365(11): 993-1003.

[515] ZAIDAT O O, FITZSIMMONS B F, WOODWARD B K, et al. Effect of a

balloon-expandable intracranial stent vs medical therapy on risk of stroke in patients

with symptomatic intracranial stenosis: the VISSIT randomized clinical trial. Jama,

2015, 313(12): 1240-1248.

[516] CHIMOWITZ M I, LYNN M J, HOWLETT-SMITH H, et al. Comparison of

warfarin and aspirin for symptomatic intracranial arterial stenosis. N Engl J Med,

2005, 352(13): 1305-1316.

[517] LIU L, WONG K S, LENG X, et al. Dual antiplatelet therapy in stroke and

ICAS: Subgroup analysis of CHANCE. Neurology, 2015, 85(13): 1154-1162.

[518] EIKELBOOM J W, CONNOLLY S J, BOSCH J, et al. Rivaroxaban with or

without Aspirin in Stable Cardiovascular Disease. N Engl J Med, 2017, 377(14):

1319-1330.

[519] PATY P S, BERNARDINI G L, MEHTA M, et al. Standardized protocols

enable stroke recognition and early treatment of carotid stenosis. J Vasc Surg, 2014,

60(1): 85-91.

[520] BARBETTA I, CARMO M, MERCANDALLI G, et al. Outcomes of urgent

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 255

carotid endarterectomy for stable and unstable acute neurologic deficits. J Vasc Surg,

2014, 59(2): 440-446.

[521] FERRERO E, FERRI M, VIAZZO A, et al. A retrospective study on early

carotid endarterectomy within 48 hours after transient ischemic attack and stroke in

evolution. Annals of vascular surgery, 2014, 28(1): 227-238.

[522] PATY P S, DARLING R C, 3RD, FEUSTEL P J, et al. Early carotid

endarterectomy after acute stroke. Journal of vascular surgery, 2004, 39(1): 148-154.

[523] LEE S B, HUH P W, KIM D S, et al. Early superficial temporal artery to middle

cerebral artery bypass in acute ischemic stroke. Clinical neurology and neurosurgery,

2013, 115(8): 1238-1244.

[524] Secondary prevention in non-rheumatic atrial fibrillation after transient

ischaemic attack or minor stroke. EAFT (European Atrial Fibrillation Trial) Study

Group. Lancet, 1993, 342(8882): 1255-1262.

[525] NG K K H, WHITELEY W. Anticoagulation Timing for Atrial Fibrillation in

Acute Ischemic Stroke: Time to Reopen Pandora's Box? JAMA neurology, 2017,

74(10): 1174-1175.

[526] PACIARONI M, AGNELLI G, FALOCCI N, et al. Early recurrence and

cerebral bleeding in patients with acute ischemic stroke and atrial fibrillation: Effect

of anticoagulation and its timing: The RAF study. Stroke, 2015, 46(8): 2175-2182.

[527] GIOIA L C, KATE M, SIVAKUMAR L, et al. Early rivaroxaban use after

cardioembolic stroke may not result in haemorrhagic transformation: A prospective

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 256

magnetic resonance imaging study. Stroke, 2016, 47(7): 1917-1919.

[528] Risk factors for stroke and efficacy of antithrombotic therapy in atrial

fibrillation. Analysis of pooled data from five randomized controlled trials. Archives

of internal medicine, 1994, 154(13): 1449-1457.

[529] LANSBERG M G, O'DONNELL M J, KHATRI P, et al. Antithrombotic and

thrombolytic therapy for ischemic stroke: Antithrombotic Therapy and Prevention of

Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical

Practice Guidelines. Chest, 2012, 141(2 Suppl): e601S-e636S.

[530] CONNOLLY S J, EIKELBOOM J, JOYNER C, et al. Apixaban in patients with

atrial fibrillation. N Engl J Med, 2011, 364(9): 806-817.

[531] CONNOLLY S J, EZEKOWITZ M D, YUSUF S, et al. Dabigatran versus

warfarin in patients with atrial fibrillation. N Engl J Med, 2009, 361(12): 1139-1151.

[532] GIUGLIANO R P, RUFF C T, BRAUNWALD E, et al. Edoxaban versus

warfarin in patients with atrial fibrillation. N Engl J Med, 2013, 369(22): 2093-2104.

[533] GRANGER C B, ALEXANDER J H, MCMURRAY J J, et al. Apixaban versus

warfarin in patients with atrial fibrillation. N Engl J Med, 2011, 365(11): 981-992.

[534] PATEL M R, MAHAFFEY K W, GARG J, et al. Rivaroxaban versus warfarin

in nonvalvular atrial fibrillation. N Engl J Med, 2011, 365(10): 883-891.

[535] AL-SADAT A, SUNBULLI M, CHATURVEDI S. Use of intravenous heparin

by north American neurologists: Do the data matter? Stroke, 2002, 33(6): 1574-1577.

[536] SCHMIDT W P, HEUSCHMANN P, TAEGER D, et al. Determinants of IV

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 257

heparin treatment in patients with ischemic stroke. Neurology, 2004, 63(12): 2407-

2409.

[537] ADAMS H P, DEL ZOPPO G, ALBERTS M J, et al. Guidelines for the early

management of adults with ischemic stroke. Stroke, 2007, 38(20): 1655-1711.

[538] ADAMS H P, ADAMS R J, BROTT T, et al. Guidelines for the early

management of patients with ischemic stroke: A scientific statement from the Stroke

Council of the American Stroke Association [M]. 2003: 1056-1083.

[539] ADAMS H P, BROTT T G, CROWELL R M, et al. Guidelines for the

management of patients with acute ischemic stroke: A statement for healthcare

professionals from a special writing group of the stroke council, american heart

association. Stroke, 1994, 25(9): 1901-1914.

[540] PAPER C. Guidelines for management of ischaemic stroke and transient

ischaemic attack 2008. Cerebrovascular diseases (Basel, Switzerland), 2008, 25(5):

457-507.

[541] COULL B M, WILLIAMS L S, GOLDSTEIN L B, et al. Anticoagulants and

antiplatelet agents in acute ischemic stroke: Report of the Joint Stroke Guideline

Development Committee of the American Academy of Neurology and the American

Stroke Association (a Division of the American Heart Association). Neurology, 2002,

59(1): 13-22.

[542] WHITELEY W N, ADAMS H P, BATH P M W, et al. Targeted use of heparin,

heparinoids, or low-molecular-weight heparin to improve outcome after acute

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 258

ischaemic stroke: an individual patient data meta-analysis of randomised controlled

trials. Lancet neurology, 2013, 12(6): 539-545.

[543] SANDERCOCK P A G, COUNSELL C, KAMAL A K. Anticoagulants for

acute ischaemic stroke. Cochrane database of systematic reviews (Online), 2008, 4):

CD000024-CD000024.

[544] YI X, LIN J, WANG C, et al. Low-molecular-weight heparin is more effective

than aspirin in preventing early neurologic deterioration and improving six-month

outcome. Journal of Stroke and Cerebrovascular Diseases, 2014, 23(6): 1537-1544.

[545] INVESTIGATORS. T P C F T T O O I A S T T. Low molecular weight

heparinoid, ORG 10172 (danaparoid), and outcome after acute ischemic stroke: a

randomized controlled trial. . Jama, 1998, 279(16): 1265-1272.

[546] LOVETT J K, COULL A J, ROTHWELL P M. Early risk of recurrence by

subtype of ischemic stroke in population-based incidence studies. Neurology, 2004,

62(4): 569-573.

[547] WONG K S, CHEN C, NG P W, et al. Low-molecular-weight heparin compared

with aspirin for the treatment of acute ischaemic stroke in Asian patients with large

artery occlusive disease: a randomised study. Lancet Neurology, 2007, 6(5): 407-413.

[548] WOESSNER R, GRAUER M, BIANCHI O, et al. Treatment with

anticoagulants in cerebral events (TRACE). Thrombosis and Haemostasis, 2004,

91(4): 690-693.

[549] MOKIN M, KASS-HOUT T, KASS-HOUT O, et al. Intravenous heparin for the

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 259

treatment of intraluminal thrombus in patients with acute ischemic stroke: A case

series. Journal of NeuroInterventional Surgery, 2013, 5(2): 144-150.

[550] VELLIMANA A K, KADKHODAYAN Y, RICH K M, et al. Symptomatic

patients with intraluminal carotid artery thrombus: outcome with a strategy of initial

anticoagulation. Journal of Neurosurgery, 2013, 118(1): 34-41.

[551] ANTITHROMBOTIC TRIALISTS C. Collaborative meta-analysis of

randomised trials of antiplatelet therapy for prevention of death, myocardial

infarction, and stroke in high risk patients. BMJ, 2002, 324(7329): 71-86.

[552] MOSER M, BODE C, NITSCHMANN S. [Clopidogrel and aspirin in patients

with atrial fibrillation : ACTIVE A study (Atrial fibrillation Clopidogrel Trial with

Irbesartan for prevention of Vascular Events).]. Der Internist, 2010, 51(1): 100-102.

[553] SLOAN M A. Clopidogrel plus aspirin was inferior to oral anticoagulation for

preventing vascular events in atrial fibrillation. Evid Based Med, 2006, 11(6): 170.

[554] FLINT A C, BANKI N M, REN X, et al. Detection of paroxysmal atrial

fibrillation by 30-day event monitoring in cryptogenic ischemic stroke: the Stroke and

Monitoring for PAF in Real Time (SMART) Registry. Stroke, 2012, 43(10): 2788-

2790.

[555] GLADSTONE D J, DORIAN P, SPRING M, et al. Atrial premature beats

predict atrial fibrillation in cryptogenic stroke: results from the EMBRACE trial.

Stroke, 2015, 46(4): 936-941.

[556] LECHAT P, MAS J L, LASCAULT G, et al. Prevalence of patent foramen ovale

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 260

in patients with stroke. N Engl J Med, 1988, 318(18): 1148-1152.

[557] HOMMA S, SACCO R L, DI TULLIO M R, et al. Effect of medical treatment

in stroke patients with patent foramen ovale: patent foramen ovale in Cryptogenic

Stroke Study. Circulation, 2002, 105(22): 2625-2631.

[558] DI TULLIO M R, SACCO R L, SCIACCA R R, et al. Patent foramen ovale and

the risk of ischemic stroke in a multiethnic population. J Am Coll Cardiol, 2007,

49(7): 797-802.

[559] KIZER J R, DEVEREUX R B. Clinical practice. Patent foramen ovale in young

adults with unexplained stroke. N Engl J Med, 2005, 353(22): 2361-2372.

[560] GU X, HE Y, LI Z, et al. Comparison of frequencies of patent foramen ovale

and thoracic aortic atherosclerosis in patients with cryptogenic ischemic stroke

undergoing transesophageal echocardiography. The American journal of cardiology,

2011, 108(12): 1815-1819.

[561] OVERELL J R, BONE I, LEES K R. Interatrial septal abnormalities and stroke:

a meta-analysis of case-control studies. Neurology, 2000, 55(8): 1172-1179.

[562] O'GARA P T, MESSE S R, TUZCU E M, et al. Percutaneous device closure of

patent foramen ovale for secondary stroke prevention: a call for completion of

randomized clinical trials: a science advisory from the American Heart

Association/American Stroke Association and the American College of Cardiology

Foundation. Circulation, 2009, 119(20): 2743-2747.

[563] NG P Y, NG A K, SUBRAMANIAM B, et al. Association of Preoperatively

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 261

Diagnosed Patent Foramen Ovale With Perioperative Ischemic Stroke. Jama, 2018,

319(5): 452-462.

[564] NAKANISHI K, YOSHIYAMA M, HOMMA S. Patent foramen ovale and

cryptogenic stroke. Trends Cardiovasc Med, 2017, 27(8): 575-581.

[565] LIBERMAN A L, DARUWALLA V J, COLLINS J D, et al. Diagnostic yield of

pelvic magnetic resonance venography in patients with cryptogenic stroke and patent

foramen ovale. Stroke, 2014, 45(8): 2324-2329.

[566] SAVER J L, CARROLL J D, THALER D E, et al. Long-Term Outcomes of

Patent Foramen Ovale Closure or Medical Therapy after Stroke. N Engl J Med, 2017,

377(11): 1022-1032.

[567] SONDERGAARD L, KASNER S E, RHODES J F, et al. Patent Foramen Ovale

Closure or Antiplatelet Therapy for Cryptogenic Stroke. N Engl J Med, 2017,

377(11): 1033-1042.

[568] MARKUS H S, HAYTER E, LEVI C, et al. Antiplatelet treatment compared

with anticoagulation treatment for cervical artery dissection (CADISS): A randomised

trial. The Lancet Neurology, 2015, 14(4): 361-367.

[569] LARSSON S C, KING A, MADIGAN J, et al. Prognosis of carotid dissecting

aneurysms: Results from CADISS and a systematic review. Neurology, 2017, 88(7):

646-652.

[570] LIP G Y, FRISON L, HALPERIN J L, et al. Comparative validation of a novel

risk score for predicting bleeding risk in anticoagulated patients with atrial

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 262

fibrillation: the HAS-BLED (Hypertension, Abnormal Renal/Liver Function, Stroke,

Bleeding History or Predisposition, Labile INR, Elderly, Drugs/Alcohol

Concomitantly) score. J Am Coll Cardiol, 2011, 57(2): 173-180.

[571] BATH P M W, WOODHOUSE L, SCUTT P, et al. Efficacy of nitric oxide, with

or without continuing antihypertensive treatment, for management of high blood

pressure in acute stroke (ENOS): a partial-factorial randomised controlled trial.

Lancet, 2015, 385(9968): 617-628.

[572] Chinese Society of Neurology, Group of Cerebrovascular Diseases, Neurology

Branch of Chinese Medical Association. Chinese Guidelines for Secondary

Prevention of Ischemic Stroke and Transient Ischemic Attack 2014 . Chinese Journal

of Neurology, 2015, 4): 258-273.

[573] HE J, ZHANG Y, XU T, et al. Effects of immediate blood pressure reduction on

death and major disability in patients with acute ischemic stroke: the CATIS

randomized clinical trial. Jama, 2014, 311(5): 479-489.

[574] XU T, ZHANG Y, BU X, et al. Blood pressure reduction in acute ischemic

stroke according to time to treatment: a subgroup analysis of the China

Antihypertensive Trial in Acute Ischemic Stroke trial. Journal of hypertension, 2017,

35(6): 1244-1251.

[575] BERGE E, COHEN G, LINDLEY R I, et al. Effects of Blood Pressure and

Blood Pressure-Lowering Treatment During the First 24 Hours Among Patients in the

Third International Stroke Trial of Thrombolytic Treatment for Acute Ischemic

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 263

Stroke. Stroke, 2015, 46(12): 3362-3369.

[576] WU W, HUO X, ZHAO X, et al. Relationship between Blood Pressure and

Outcomes in Acute Ischemic Stroke Patients Administered Lytic Medication in the

TIMS-China Study. PloS one, 2016, 11(2): e0144260.

[577] JI N, MENG P, LIU N, et al. A Reasonable Blood Pressure Level for Good

Clinical Outcome After the Acute Phase of Ischemic Stroke. Journal of clinical

hypertension (Greenwich, Conn), 2016, 18(6): 536-542.

[578] KIRK J K, ALLSBROOK J, HANSELL M, et al. A systematic review of

hypertension outcomes and treatment strategies in older adults. Archives of

Gerontology & Geriatrics, 2017, 73(160-168.

[579] BATH P M, MARTIN R H, PALESCH Y, et al. Effect of telmisartan on

functional outcome, recurrence, and blood pressure in patients with acute mild

ischemic stroke: a PRoFESS subgroup analysis. Stroke, 2009, 40(11): 3541-3546.

[580] LEE M, OVBIAGELE B, HONG K S, et al. Effect of Blood Pressure Lowering

in Early Ischemic Stroke: Meta-Analysis. Stroke, 2015, 46(7): 1883-1889.

[581] ZHAO R, LIU F D, WANG S, et al. Blood Pressure Reduction in the Acute

Phase of an Ischemic Stroke Does Not Improve Short- or Long-Term Dependency or

Mortality: A Meta-Analysis of Current Literature. Medicine, 2015, 94(23): e896.

[582] BU X, LI C, ZHANG Y, et al. Early Blood Pressure Reduction in Acute

Ischemic Stroke with Various Severities: A Subgroup Analysis of the CATIS Trial.

Cerebrovascular diseases (Basel, Switzerland), 2016, 42(3-4): 186-195.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 264

[583] WILLMOT M, GHADAMI A, WHYSALL B, et al. Transdermal glyceryl

trinitrate lowers blood pressure and maintains cerebral blood flow in recent stroke.

Hypertension, 2006, 47(6): 1209-1215.

[584] NAKAMURA T, TSUTSUMI Y, SHIMIZU Y, et al. Renin-angiotensin system

blockade safely reduces blood pressure in patients with minor ischemic stroke during

the acute phase. Journal of stroke and cerebrovascular diseases : the official journal of

National Stroke Association, 2010, 19(6): 435-440.

[585] OH M S, YU K H, HONG K S, et al. Modest blood pressure reduction with

valsartan in acute ischemic stroke: a prospective, randomized, open-label, blinded-

end-point trial. International journal of stroke : official journal of the International

Stroke Society, 2015, 10(5): 745-751.

[586] ZHAI Z Y, DU C Y, SUN M, et al. Association of isolated diastolic

hypertension with carotid artery plaque ulceration in transient ischemic attack patients

under the age of 50. Journal of Clinical Neurology, 2014, 6): 413-415.

[587] SANDSET E C, JUSUFOVIC M, SANDSET P M, et al. Effects of blood

pressure-lowering treatment in different subtypes of acute ischemic stroke. Stroke,

2015, 46(3): 877-879.

[588] WOHLFAHRT P, KRAJCOVIECHOVA A, JOZIFOVA M, et al. Low blood

pressure during the acute period of ischemic stroke is associated with decreased

survival. Journal of Hypertension, 2015, 33(2): 339-345.

[589] VEMMOS K N, TSIVGOULIS G, SPENGOS K, et al. U-shaped relationship

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 265

between mortality and admission blood pressure in patients with acute stroke. Journal

of Internal Medicine, 2004, 255(2): 257-265.

[590] OKUMURA K, OHYA Y, MAEHARA A, et al. Effects of blood pressure levels

on case fatality after acute stroke. Journal of Hypertension, 2005, 23(6): 1217-1223.

[591] STEAD L G, GILMORE R M, DECKER W W, et al. Initial emergency

department blood pressure as predictor of survival after acute ischemic stroke.

Neurology, 2005, 65(8): 1179-1183.

[592] LEONARDI-BEE J, BATH P M W, PHILLIPS S J, et al. Blood pressure and

clinical outcomes in the international stroke trial. Stroke, 2002, 33(5): 1315-1320.

[593] MANNING L S, MISTRI A K, POTTER J, et al. Short-Term Blood Pressure

Variability in Acute Stroke Post Hoc Analysis of the Controlling Hypertension and

Hypotension Immediately Post Stroke and Continue or Stop Post-Stroke

Antihypertensives Collaborative Study Trials. Stroke, 2015, 46(6): 1518-+.

[594] MUSCARI A, PUDDU G M, SERAFINI C, et al. Predictors of short-term

improvement of ischemic stroke. Neurological Research, 2013, 35(6): 594-601.

[595] VISVANATHAN A, DENNIS M, WHITELEY W. Parenteral fluid regimens for

improving functional outcome in people with acute stroke. Cochrane Database of

Systematic Reviews, 2015, 9):

[596] HORN J, DE HAAN R J, VERMEULEN M, et al. Very early nimodipine use in

stroke (VENUS) - A randomized, double-blind, placebo-controlled trial. Stroke, 2001,

32(2): 461-465.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 266

[597] SCHRADER J, LUDERS S, KULSCHEWSKI A, et al. The ACCESS study -

Evaluation of acute candesartan cilexetil therapy in stroke survivors. Stroke, 2003,

34(7): 1699-1703.

[598] POTTER J F, ROBINSON T G, FORD G A, et al. Controlling hypertension and

hypotension immediately post-stroke (CHHIPS): a randomised, placebo-controlled,

double-blind pilot trial. Lancet Neurology, 2009, 8(1): 48-56.

[599] ROBINSON T G, POTTER J F, FORD G A, et al. Effects of antihypertensive

treatment after acute stroke in the Continue Or Stop post-Stroke Antihypertensives

Collaborative Study (COSSACS): a prospective, randomised, open, blinded-endpoint

trial. Lancet Neurology, 2010, 9(8): 767-775.

[600] HE J, ZHANG Y H, XU T, et al. Effects of Immediate Blood Pressure

Reduction on Death and Major Disability in Patients With Acute Ischemic Stroke The

CATIS Randomized Clinical Trial. Jama-J Am Med Assoc, 2014, 311(5): 479-489.

[601] GROUP P C. Post-stroke antihypertensive treatment study. A preliminary result.

Chin Med J (Engl), 1995, 108(9): 710-717.

[602] WEINBERGER J. Randomised trial of a perindopril-based blood-pressure-

lowering regimen among 6105 individuals with previous stroke or transient ischaemic

attack. Curr Cardiol Rep, 2003, 5(2): 140.

[603] LIU L, WANG Z, GONG L, et al. Blood pressure reduction for the secondary

prevention of stroke: a Chinese trial and a systematic review of the literature.

Hypertens Res, 2009, 32(11): 1032-1040.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 267

[604] ROBINSON T G, POTTER J F, FORD G A, et al. Effects of antihypertensive

treatment after acute stroke in the Continue or Stop Post-Stroke Antihypertensives

Collaborative Study (COSSACS): a prospective, randomised, open, blinded-endpoint

trial. Lancet Neurol, 2010, 9(8): 767-775.

[605] HE J, ZHANG Y, XU T, et al. Effects of immediate blood pressure reduction on

death and major disability in patients with acute ischemic stroke: the CATIS

randomized clinical trial. JAMA, 2014, 311(5): 479-489.

[606] CHIMOWITZ M I, LYNN M J, DERDEYN C P, et al. Stenting versus

aggressive medical therapy for intracranial arterial stenosis. N Engl J Med, 2011,

365(11): 993-1003.

[607] FORSTER A, SZABO K, HENNERICI M G. Pathophysiological concepts of

stroke in hemodynamic risk zones--do hypoperfusion and embolism interact? Nat

Clin Pract Neurol, 2008, 4(4): 216-225.

[608] PERGOLA P E, WHITE C L, SZYCHOWSKI J M, et al. Achieved blood

pressures in the secondary prevention of small subcortical strokes (SPS3) study:

challenges and lessons learned. Am J Hypertens, 2014, 27(8): 1052-1060.

[609] DRAWZ P E, PAJEWSKI N M, BATES J T, et al. Effect of Intensive Versus

Standard Clinic-Based Hypertension Management on Ambulatory Blood Pressure:

Results From the SPRINT (Systolic Blood Pressure Intervention Trial) Ambulatory

Blood Pressure Study. Hypertension, 2017, 69(1): 42-50.

[610] MANCIA G. Effects of intensive blood pressure control in the management of

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 268

patients with type 2 diabetes mellitus in the Action to Control Cardiovascular Risk in

Diabetes (ACCORD) trial. Circulation, 2010, 122(8): 847-849.

[611] HUISA B N, STEMER A B, ZIVIN J A. Atorvastatin in stroke: a review of

SPARCL and subgroup analysis. Vascular health and risk management, 2010, 6(229-

236.

[612] STONE N J, ROBINSON J G, LICHTENSTEIN A H, et al. 2013 ACC/AHA

guideline on the treatment of blood cholesterol to reduce atherosclerotic

cardiovascular risk in adults: a report of the American College of

Cardiology/American Heart Association Task Force on Practice Guidelines. J Am

Coll Cardiol, 2014, 63(25 Pt B): 2889-2934.

[613] YOSHIMURA S, UCHIDA K, DAIMON T, et al. Randomized Controlled Trial

of Early Versus Delayed Statin Therapy in Patients With Acute Ischemic Stroke:

ASSORT Trial (Administration of Statin on Acute Ischemic Stroke Patient). Stroke,

2017, 48(11): 3057-3063.

[614] LEE V W, CHAU R Y, CHEUNG H Y, et al. How low should we target the

LDL goal to improve survival for acute coronary syndrome patients in Hong Kong?

BMC cardiovascular disorders, 2015, 15(117.

[615] LEIBOWITZ M, COHEN-STAVI C, BASU S, et al. Targeting LDL

Cholesterol: Beyond Absolute Goals Toward Personalized Risk. Current cardiology

reports, 2017, 19(6): 52.

[616] ANDERSON T J, GREGOIRE J, HEGELE R A, et al. 2012 update of the

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 269

Canadian Cardiovascular Society guidelines for the diagnosis and treatment of

dyslipidemia for the prevention of cardiovascular disease in the adult. Can J Cardiol,

2013, 29(2): 151-167.

[617] KERNAN W N, OVBIAGELE B, BLACK H R, et al. Guidelines for the

prevention of stroke in patients with stroke and transient ischemic attack: a guideline

for healthcare professionals from the American Heart Association/American Stroke

Association. Stroke, 2014, 45(7): 2160-2236.

[618] SILLESEN H, AMARENCO P, HENNERICI M G, et al. Atorvastatin reduces

the risk of cardiovascular events in patients with carotid atherosclerosis: a secondary

analysis of the Stroke Prevention by Aggressive Reduction in Cholesterol Levels

(SPARCL) trial. Stroke, 2008, 39(12): 3297-3302.

[619] TURAN T N, NIZAM A, LYNN M J, et al. Relationship between risk factor

control and vascular events in the SAMMPRIS trial. Neurology, 2017, 88(4): 379-

385.

[620] FLINT A C, CONELL C, REN X, et al. Statin Adherence Is Associated With

Reduced Recurrent Stroke Risk in Patients With or Without Atrial Fibrillation. Stroke,

2017, 48(7): 1788-1794.

[621] KOSKINAS K C, SIONTIS G C M, PICCOLO R, et al. Effect of statins and

non-statin LDL-lowering medications on cardiovascular outcomes in secondary

prevention: a meta-analysis of randomized trials. European heart journal, 2017,

[622] SABATINE M S, GIUGLIANO R P, KEECH A C, et al. Evolocumab and

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 270

Clinical Outcomes in Patients with Cardiovascular Disease. The New England journal

of medicine, 2017, 376(18): 1713-1722.

[623] SCHANDELMAIER S, BRIEL M, SACCILOTTO R, et al. Niacin for primary

and secondary prevention of cardiovascular events. The Cochrane database of

systematic reviews, 2017, 6(Cd009744.

[624] The DELP study group. Initial Analysis of Multicenter Randomized Controlled

Clinical Trial of DELP System in Treating Patients with Acute Cerebral Infarction.

Neural Injury and Functional Reconstruction, 2008, 3(1): 8-11.

[625] SHARP COLLABORATIVE G. Study of Heart and Renal Protection (SHARP):

randomized trial to assess the effects of lowering low-density lipoprotein cholesterol

among 9,438 patients with chronic kidney disease. American heart journal, 2010,

160(5): 785-794.e710.

[626] ROSSEBO A B, PEDERSEN T R, BOMAN K, et al. Intensive lipid lowering

with simvastatin and ezetimibe in aortic stenosis. N Engl J Med, 2008, 359(13): 1343-

1356.

[627] MURPHY S A, CANNON C P, BLAZING M A, et al. Reduction in Total

Cardiovascular Events With Ezetimibe/Simvastatin Post-Acute Coronary Syndrome:

The IMPROVE-IT Trial. J Am Coll Cardiol, 2016, 67(4): 353-361.

[628] OLAIYA M T, CADILHAC D A, KIM J, et al. Community-Based Intervention

to Improve Cardiometabolic Targets in Patients With Stroke: A Randomized

Controlled Trial. Stroke, 2017, 48(9): 2504-2510.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 271

[629] KENNEDY J, HILL M D, RYCKBORST K J, et al. Fast assessment of stroke

and transient ischaemic attack to prevent early recurrence (FASTER): a randomised

controlled pilot trial. The Lancet Neurology, 2007, 6(11): 961-969.

[630] SANOSSIAN N, SAVER J L, LIEBESKIND D S, et al. Achieving target

cholesterol goals after stroke: is in-hospital statin initiation the key? Archives of

neurology, 2006, 63(8): 1081-1083.

[631] LEE M, SAVER J L, WU Y L, et al. Utilization of Statins Beyond the Initial

Period After Stroke and 1-Year Risk of Recurrent Stroke. Journal of the American

Heart Association, 2017, 6(8):

[632] CATAPANO A L, GRAHAM I, DE BACKER G, et al. 2016 ESC/EAS

Guidelines for the Management of Dyslipidaemias. European heart journal, 2016,

37(39): 2999-3058.

[633] CHATURVEDI S, ZIVIN J, BREAZNA A, et al. Effect of atorvastatin in

elderly patients with a recent stroke or transient ischemic attack. Neurology, 2009,

72(8): 688-694.

[634] O'REGAN C, WU P, ARORA P, et al. Statin therapy in stroke prevention: a

meta-analysis involving 121,000 patients. The American journal of medicine, 2008,

121(1): 24-33.

[635] KATO E T, CANNON C P, BLAZING M A, et al. Efficacy and Safety of

Adding Ezetimibe to Statin Therapy Among Women and Men: Insight From

IMPROVE-IT (Improved Reduction of Outcomes: Vytorin Efficacy International

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 272

Trial). Journal of the American Heart Association, 2017, 6(11):

[636] GIUGLIANO R P, CANNON C P, BLAZING M A, et al. Benefit of Adding

Ezetimibe to Statin Therapy on Cardiovascular Outcomes and Safety in Patients With

vs. Without Diabetes: Results from IMPROVE-IT. Circulation, 2017,

[637] BOHULA E A, WIVIOTT S D, GIUGLIANO R P, et al. Prevention of Stroke

with the Addition of Ezetimibe to Statin Therapy in Patients With Acute Coronary

Syndrome in IMPROVE-IT (Improved Reduction of Outcomes: Vytorin Efficacy

International Trial). Circulation, 2017, 136(25): 2440-2450.

[638] PALACIO E, VIADERO-CERVERA R, REVILLA M, et al. [Utility of

treatment with atorvastatin 40 mg plus ezetimibe 10 mg versus atorvastatin 80 mg in

reducing the levels of LDL cholesterol in patients with ischaemic stroke or transient

ischaemic attack]. Revista de neurologia, 2016, 62(5): 203-210.

[639] GENTILE N T, SEFTCHICK M W, HUYNH T, et al. Decreased mortality by

normalizing blood glucose after acute ischemic stroke. Acad Emerg Med, 2006, 13(2):

174-180.

[640] WILLIAMS L S, ROTICH J, QI R, et al. Effects of admission hyperglycemia

on mortality and costs in acute ischemic stroke. Neurology, 2002, 59(1): 67-71.

[641] CAPES S E, HUNT D, MALMBERG K, et al. Stress hyperglycemia and

prognosis of stroke in nondiabetic and diabetic patients: a systematic overview.

Stroke, 2001, 32(10): 2426-2432.

[642] MCCORMICK M T, MUIR K W, GRAY C S, et al. Management of

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 273

hyperglycemia in acute stroke: how, when, and for whom? Stroke, 2008, 39(7): 2177-

2185.

[643] BRUNO A, LEVINE S R, FRANKEL M R, et al. Admission glucose level and

clinical outcomes in the NINDS rt-PA Stroke Trial. Neurology, 2002, 59(5): 669-674.

[644] DEMCHUK A M, TANNE D, HILL M D, et al. Predictors of good outcome

after intravenous tPA for acute ischemic stroke. Neurology, 2001, 57(3): 474-480.

[645] ELS T, KLISCH J, ORSZAGH M, et al. Hyperglycemia in patients with focal

cerebral ischemia after intravenous thrombolysis: influence on clinical outcome and

infarct size. Cerebrovasc Dis, 2002, 13(2): 89-94.

[646] PARSONS M W, BARBER P A, DESMOND P M, et al. Acute hyperglycemia

adversely affects stroke outcome: a magnetic resonance imaging and spectroscopy

study. Annals of neurology, 2002, 52(1): 20-28.

[647] RIBO M, MOLINA C A, DELGADO P, et al. Hyperglycemia during ischemia

rapidly accelerates brain damage in stroke patients treated with tPA. Journal of

cerebral blood flow and metabolism : official journal of the International Society of

Cerebral Blood Flow and Metabolism, 2007, 27(9): 1616-1622.

[648] GRAY C S, HILDRETH A J, SANDERCOCK P A, et al. Glucose-potassium-

insulin infusions in the management of post-stroke hyperglycaemia: the UK Glucose

Insulin in Stroke Trial (GIST-UK). The Lancet Neurology, 2007, 6(5): 397-406.

[649] BRUNO A, KENT T A, COULL B M, et al. Treatment of hyperglycemia in

ischemic stroke (THIS): a randomized pilot trial. Stroke, 2008, 39(2): 384-389.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 274

[650] JOHNSTON K C, HALL C E, KISSELA B M, et al. Glucose Regulation in

Acute Stroke Patients (GRASP) trial: a randomized pilot trial. Stroke, 2009, 40(12):

3804-3809.

[651] KREISEL S H, BERSCHIN U M, HAMMES H P, et al. Pragmatic management

of hyperglycaemia in acute ischaemic stroke: safety and feasibility of intensive

intravenous insulin treatment. Cerebrovasc Dis, 2009, 27(2): 167-175.

[652] ACTION TO CONTROL CARDIOVASCULAR RISK IN DIABETES STUDY

G, GERSTEIN H C, MILLER M E, et al. Effects of intensive glucose lowering in

type 2 diabetes. N Engl J Med, 2008, 358(24): 2545-2559.

[653] DUCKWORTH W, ABRAIRA C, MORITZ T, et al. Glucose control and

vascular complications in veterans with type 2 diabetes. N Engl J Med, 2009, 360(2):

129-139.

[654] GROUP A C, PATEL A, MACMAHON S, et al. Intensive blood glucose control

and vascular outcomes in patients with type 2 diabetes. N Engl J Med, 2008, 358(24):

2560-2572.

[655] MARSO S P, KENNEDY K F, HOUSE J A, et al. The effect of intensive

glucose control on all-cause and cardiovascular mortality, myocardial infarction and

stroke in persons with type 2 diabetes mellitus: a systematic review and meta-

analysis. Diab Vasc Dis Res, 2010, 7(2): 119-130.

[656] GORELICK P B, RODIN M B, LANGENBERG P, et al. Weekly alcohol

consumption, cigarette smoking, and the risk of ischemic stroke: results of a case-

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 275

control study at three urban medical centers in Chicago, Illinois. Neurology, 1989,

39(3): 339-343.

[657] JAMROZIK K, BROADHURST R J, ANDERSON C S, et al. The role of

lifestyle factors in the aetiology of stroke. A population-based case-control study in

Perth, Western Australia. Stroke, 1994, 25(1): 51-59.

[658] ELLEKJAER E F, WYLLER T B, SVERRE J M, et al. Lifestyle factors and

risk of cerebral infarction. Stroke, 1992, 23(6): 829-834.

[659] CAZZATO G, ZORZON M, MONTI F, et al. [Smoking and acute

cerebrovascular disorders of the ischemic type]. Rivista di neurologia, 1985, 55(2):

88-98.

[660] SONG Y M, KWON S U, SUNG J, et al. Different risk factor profiles between

subtypes of ischemic stroke. A case-control study in Korean men. European journal of

epidemiology, 2005, 20(7): 605-612.

[661] JI R, PAN Y, YAN H, et al. Current smoking is associated with extracranial

carotid atherosclerotic stenosis but not with intracranial large artery disease. BMC

neurology, 2017, 17(1): 120.

[662] MCEVOY R D, ANTIC N A, HEELEY E, et al. CPAP for Prevention of

Cardiovascular Events in Obstructive Sleep Apnea. N Engl J Med, 2016, 375(10):

919-931.

[663] SUNDELL L, SALOMAA V, VARTIAINEN E, et al. Increased stroke risk is

related to a binge-drinking habit. Stroke, 2008, 39(12): 3179-3184.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 276

[664] MOSTOFSKY E, BURGER M R, SCHLAUG G, et al. Alcohol and acute

ischemic stroke onset: the stroke onset study. Stroke, 2010, 41(9): 1845-1849.

[665] PATRA J, TAYLOR B, IRVING H, et al. Alcohol consumption and the risk of

morbidity and mortality for different stroke types--a systematic review and meta-

analysis. BMC public health, 2010, 10(258.

[666] RANTAKOMI S H, LAUKKANEN J A, SIVENIUS J, et al. Alcohol

consumption and the risk of stroke among hypertensive and overweight men. Journal

of neurology, 2013, 260(2): 534-539.

[667] JONES S B, LOEHR L, AVERY C L, et al. Midlife Alcohol Consumption and

the Risk of Stroke in the Atherosclerosis Risk in Communities Study. Stroke, 2015,

46(11): 3124-3130.

[668] LARSSON S C, WALLIN A, WOLK A, et al. Differing association of alcohol

consumption with different stroke types: a systematic review and meta-analysis. BMC

medicine, 2016, 14(1): 178.

[669] IKEHARA S, ISO H, TOYOSHIMA H, et al. Alcohol consumption and

mortality from stroke and coronary heart disease among Japanese men and women:

the Japan collaborative cohort study. Stroke, 2008, 39(11): 2936-2942.

[670] ANGEJA B G, SHLIPAK M G, GO A S, et al. Hormone therapy and the risk of

stroke after acute myocardial infarction in postmenopausal women. Journal of the

American College of Cardiology, 2001, 38(5): 1297-1301.

[671] BUSHNELL C D, SAMSA G P, GOLDSTEIN L B. Hormone replacement

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 277

therapy and ischemic stroke severity in women: a case-control study. Neurology,

2001, 56(10): 1304-1307.

[672] BUSHNELL C. Stroke Hormones and Outcomes in Women (SHOW) study: is

the 'healthy-user effect' valid for women after stroke? Womens Health, 2009, 5(5):

485-496.

[673] CHEETHAM T C, AN J, JACOBSEN S J, et al. Association of Testosterone

Replacement With Cardiovascular Outcomes Among Men With Androgen Deficiency.

Jama Intern Med, 2017, 177(4): 491-499.

[674] SIDNEY S, CHEETHAM T C, CONNELL F A, et al. Recent combined

hormonal contraceptives (CHCs) and the risk of thromboembolism and other

cardiovascular events in new users. Contraception, 2013, 87(1): 93-100.

[675] CHEN J F, LI Y, ZHOU J W, et al. A study of relationship between oral

contraceptives and gene polymorphism and types of stroke. Chinese Journal of

Epidemiology, 2001, 04): 39-42.

[676] CHEN T, LI Y, WANG C, et al. The joint effects of hypertension and low-dose

of COC on the risk of stroke in women. Chinese Journal of Disease Control &

Prevention, 2014, 02): 135-138.

[677] SHEN H, WANG M Z, PENG Z Q, et al. Correlation between application of

compound oral contraceptives and cerebrovascular disease in women of childbearing

age in Beijing. Chinese Journal of Family Planning, 1995, 05): 264-268+320.

[678] SUN Z M, LI Y. Risk Factors for Haemorrhagic Stroke and Oral Contraceptives

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 278

among Chinese Women:1:1 Case-control Study. Chinese Journal of Family Planning,

2004, 10): 606-609.

[679] CHENG Y C, RYAN K A, QADWAI S A, et al. Cocaine Use and Risk of

Ischemic Stroke in Young Adults. Stroke, 2016, 47(4): 918-922.

[680] GIRALDO E A, TAQI M A, VAIDEAN G D. A case-control study of stroke

risk factors and outcomes in African American stroke patients with and without crack-

cocaine abuse. Neurocritical care, 2012, 16(2): 273-279.

[681] SLOAN M A, KITTNER S J, FEESER B R, et al. Illicit drug-associated

ischemic stroke in the Baltimore-Washington Young Stroke Study. Neurology, 1998,

50(6): 1688-1693.

[682] BHATTACHARYA P, TARAMAN S, SHANKAR L, et al. Clinical profiles,

complications, and disability in cocaine-related ischemic stroke. Journal of stroke and

cerebrovascular diseases : the official journal of National Stroke Association, 2011,

20(5): 443-449.

[683] WALD D S, LAW M, MORRIS J K. Homocysteine and cardiovascular disease:

evidence on causality from a meta-analysis. BMJ, 2002, 325(7374): 1202.

[684] HOMOCYSTEINE STUDIES C. Homocysteine and risk of ischemic heart

disease and stroke: a meta-analysis. Jama, 2002, 288(16): 2015-2022.

[685] HOLMES M V, NEWCOMBE P, HUBACEK J A, et al. Effect modification by

population dietary folate on the association between MTHFR genotype,

homocysteine, and stroke risk: a meta-analysis of genetic studies and randomised

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 279

trials. Lancet, 2011, 378(9791): 584-594.

[686] SPENCE J D, BANG H, CHAMBLESS L E, et al. Vitamin Intervention For

Stroke Prevention trial: an efficacy analysis. Stroke, 2005, 36(11): 2404-2409.

[687] GROUP V T S. B vitamins in patients with recent transient ischaemic attack or

stroke in the VITAmins TO Prevent Stroke (VITATOPS) trial: a randomised, double-

blind, parallel, placebo-controlled trial. The Lancet Neurology, 2010, 9(9): 855-865.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 280

Supplemental Table 1 Flow chart of skull imagological examination for patients

suspected of ischemic stroke after entering emergency department

All patients suspected of acute stroke

Time point Imaging Main assessment content

examination

Within 30min after entry into NCCT Exclusion of haemorrhage

emergency department ASPECTS ≥ 6 indicates

macrovascular occlusion

Indications for intravascular treatment for suspected macrovascular occlusion

Time point Imaging Main assessment content

examination

Within 6h of onset CTA Whether there is macrovascular

occlusion

Within 16h after onset DWI/PWI or Whether compliance with DAWN or

CTP DEFUSE3 standard

Within 24h after onset DWI/PWI or Whether compliance with DAWN

CTP standard

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 281

Supplemental Table 2 Set thresholds of different imaging strategies for ischemic

penumbra

Imaging strategy Set threshold for ischemic penumbra

DWI-PWI mismatch The area with Tmax > 6s on PWI is set as the low perfusion area, the

area with ADC < 600×10-6mm2/s on DWI is set as the infarction

core area, the target mismatch is defined as infarction core area <

70ml, ischemic penumbra > 15ml, and the ratio of total low

perfusion area to ischemic core area of > 1.8

CT PWI The area with CBF lower than 30% of the normal tissue on the

affected side is set as the infarction core lesion, the area with Tmax >

6s is set as the low perfusion area, and the definition of target

mismatch is the same as before.

Clinical imaging mismatch The patients were divided into three groups according to the size,

age and NIHSS score of the imaging lesion: Group A was over 80

years old, NIHSS score ≥ 10 and infarction core < 21ml. Group B

was under 80 years old, NIHSS score ≥ 10 and infarction core <

31ml. Group C was under 80 years old, NIHSS score ≥ 20 and

infarction core of 31--51ml.

DWI-FLAIR mismatch Lesions can be seen on DWI but there is no high signal on the

corresponding parts of FLAIR

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 282

Supplemental Table 3 Indications and contraindications of rt-PA intravenous

thrombolysis within 3h

Indications

1. There are neurological impairment symptoms caused by ischemic stroke

2. Occurrence of symptoms < 3h

3. Age ≥ 18 years

4. Patients or their families sign the informed consent form

Contraindications

1. Intracranial hemorrhage (such as cerebral parenchyma hemorrhage, intraventricular

hemorrhage, subarachnoid hemorrhage, subdural / epidural hematoma)

2. History of intracranial haemorrhage

3. Severe head trauma or stroke in the past 3 months

4. Intracranial tumour, giant intracranial aneurysms

5. Recent (in 3 months) intracranial or intraspinal surgery

6. Major surgeries in the last two weeks

7. Gastrointestinal or urinary system bleeding in the last 3 weeks

8. Active internal haemorrhage

9. Aortic arch dissection

10. Arterial puncture in a site where haemostasis by compression is not easy to within the past

week

11. Elevation of blood pressure, systolic pressure ≥ 180mmHg or diastolic pressure ≥

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 283

100mmHg

12. Acute haemorrhagic tendency, including platelet count below 100× 109/L or other

conditions

13. Receipt of low molecular weight heparin treatment within 24h

14. INR > 1.7 or PT > 15s for patients who have taken anticoagulant orally

15. Thrombin inhibitors or Xa factor inhibitors are being used within 48h, or the results of

laboratory tests are abnormal (such as APTT, INR, platelet count, ECT; TT or appropriate

Xa factor activity determination)

16. Blood glucose < 2.8mmol/L or > 22.22 mmol/L

17. Head CT or MRI indicates a large area of infarction (infarct size > 1/3 the blood supply

area of middle cerebral artery)

Relative contraindications

The risks and benefits of thrombolysis should be carefully considered and weighed in the

following situations (i.e. although there are one or more relative contraindications, thrombolysis

is not absolutely impossible):

1. Minor non-disabling stroke

2. Stroke with rapid improvement of symptoms

3. Neurological impairment symptoms after epileptic seizure (associated with this stroke)

4. Extracranial cervical arterial dissection

5. Severe trauma in the past 2 weeks (no head injury)

6. History of myocardial infarction in the past 3 months

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 284

7. Pregnancy

8. Dementia

9. Severe neurological disability left by previous diseases

10. Unruptured and untreated arteriovenous malformations and small intracranial aneurysms (<

10mm)

11. A small amount of intracerebral microhemorrhage (1~10)

12. Use of prohibited drugs

13. Stroke mimics

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 285

Supplemental Table 4 Indications and contraindications of rt-PA intravenous

thrombolysis within 3--4.5h

Indications

1. There are neurological impairment symptoms caused by ischemic stroke

2. Symptom occurrence 3-4.5h

3. Age ≥ 18 years

4. Patients or their families sign the informed consent form

Contraindications

Same as Supplemental Table 3contraindications

Relative contraindications

The risks and benefits of thrombolysis should be carefully considered and weighed in the

following situations (i.e. although there are one or more relative contraindications, thrombolysis

is not absolutely impossible):

1~13 items are the same as those in Supplemental Table 3 Relative contraindications.

14. INR ≤ 1.7 and PT ≤ 15s for patients who have taken anticoagulant orally

15. Severe stroke (NIHSS score > 25)

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 286

Supplemental Table 5 Blood pressure control for thrombolysis

Other than patients with blood pressure > 185/110mmHg, for patients suitable for acute

reperfusion therapy:

Labetalol 10--20 mg is injected intravenously for 1--2 min, which may be repeated once;

Nicardipine 5mg/h intravenous injection. After every 5--15 min, the dripping speed can be

increased by 2.5mg/h, and the maximum dripping speed is 15 mg/h. When the expected blood

pressure value is reached, the dripping speed should be adjusted to maintain the ideal blood

pressure;

Clovidipine is injected intravenously at 1--2 mg/h. The dripping speed can be doubled every 2--5

minutes until the expected blood pressure is reached, with the maximum dripping speed being 21

mg/h;

Other drugs, such as hydralazine and enalapril, may also be considered.

Thrombolytic therapy must not be performed if the blood pressure is not maintained at ≤

180/110mmHg.

In the blood pressure management during thrombolysis, after thrombolysis or other acute

reperfusion therapy, the blood pressure should be maintained at ≤ 180/105mmHg.

Blood pressure should be monitored every 15min in 2h, every 30min in 6h, and once every hour in

16h after thrombolytic therapy.

If SBP > 180--230mmHg or DBP > 105--120mmHg:

Labetalol 10mg intravenous injection is followed by intravenous infusion of 2--8 mg/min;

Nicardipine 5mg/h intravenous injection. After every 5--15 min, the dripping speed can be

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 287

increased by 2.5mg/h to reach the expected blood pressure value. The maximum dropping speed is

15 mg/h;

Clovidipine is injected intravenously at 1- 2 mg/h. The dripping speed can be doubled every 2- 5

minutes until the expected blood pressure is reached, with the maximum dripping speed being 21

mg/h;

If blood pressure is not controlled or diastolic pressure is > 140mmHg, intravenous injection

of sodium nitroprusside should be considered.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 288

Supplemental Table 6 Monitoring and handling of intravenous thrombolysis

1.Patients should be admitted to a neurological intensive care unit or a stroke unit for

monitoring

2. Regular blood pressure and neurological function tests should be carried out. Blood pressure

measurements and neurological function assessments should be carried out every 15min within

2 hours after intravenous thrombolytic therapy is completed. After that, they should be carried

out every 30min for 6 hours, then once every hour until 24 hours after the end of treatment

3. In case of severe headache, hypertension, nausea or vomiting, or deterioration of

neurological symptoms and signs, thrombolytic drugs should be withdrawn immediately and

head CT examination should be performed

4.In case of SBP ≥ 180mmHg or DBP ≥ 105mmHg, the frequency of blood pressure

monitoring should be increased and antihypertensive drugs should be given

5.Placement of nasal feeding tube, urethral catheter and intra-arterial pressure tube should be

delayed when the condition permits

6.24h after thrombolytic therapy, head CT or MRI should be performed again before

antiplatelet drugs or anticoagulants are given

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 289

Supplemental Table 7 Imaging screening criteria beyond time windows in

different studies

Study name Inclusion criteria

DAWN study ① The time from the last seemingly normal time of the patient to

randomization was 6--24 h.

② The screening scheme is that the severity of clinical neurological

impairment symptoms does not match the infarct volume-"clinical-

imaging mismatch" (NIHSS score does not match the infarct volume of

MRI-DWI/CTP-rCBF), which is defined as:

Group A: ≥ 80 years, NIHSS score ≥ 10, infarct volume < 21ml

Group B: < 80 years, NIHSS score ≥ 10, infarct volume < 31ml

Group C: < 80 years, NIHSS score ≥ 20, infarct volume < 51ml

DEFUSE 3 Preoperative mRS score ≤ 2, aged 18-90 years

study The time from onset to start of intravascular treatment is 6-16h

DWI or CTP cerebral infarction core volume ≤ 70ml

Ratio of hypoperfusion area/infarction area volume ≥ 1.8 or mismatch volume

between hypoperfusion area and infarction area > 15ml

Refer to Figure 3 for details of the treatment process of intravascular treatment for

patients with acute ischemic stroke.

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 290

Supplemental Table 8 Antiplatelet drugs commonly used for ischemic stroke

Medication Target and mechanism of action Indication

plan

Aspirin Through irreversible acetylation with hydroxyl group of AIS patients should take it within 24-48h after onset. For patients treated with

residue 530 in polypeptide chain of COX-1 active site in intravenous alteplase, aspirin is usually postponed until 24h later (however, in

cyclooxygenase (COX), Cox is inactivated, thus blocking the the case of some complications, in the absence of intravenous alteplase

pathway of AA conversion to A2 (TXA2) and treatment, if it is known that administration of aspirin can bring significant

inhibiting platelet aggregation benefits or the absence of aspirin will cause significant risks, not postponing it

may be considered)

Aspirin as substitute for treatment is not recommended for acute stroke patients

who are suitable for intravenous thrombolysis with alteplase or mechanical

thrombectomy

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 291

Clopidogrel It selectively inhibits ADP binding to platelet receptors and For patients with mild stroke and TIA of medium and high risk, dual

inhibits ADP-mediated activation of glycoprotein (GP)Ⅱb/Ⅲa antiplatelet therapy (aspirin 100mg/ time, qd, combined with clopidogrel 75mg/

complex, thus inhibiting platelet aggregation. It can also inhibit time, qd, clopidogrel loading dose 300mg on the first day) should be started

platelet aggregation not caused by ADP. Its effect on platelet within 24 hours of onset, which is beneficial to prevent early stroke recurrence

ADP receptor is irreversible. Its oral absorption is rapid, the within 90 days of onset

protein binding rate in plasma is 98%, and it is metabolized in For mild stroke patients complicated with high-risk intracranial arterial stenosis

liver (70%-99%), combined stent therapy is not recommended on the basis of dual

antiplatelet therapy (aspirin combined with clopidogrel)

Ticagrelor It is a selective ADP receptor antagonist that acts on P2Y12 Ticagrelor (instead of aspirin) is not recommended for acute treatment of mild

ADP receptor to inhibit ADP-mediated platelet activation and stroke

aggregation, with similar mechanism of action of

drugs (such as clopidogrel). However, the

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 292

difference is that the interaction between ticagrelor and platelet

P2Y12 ADP receptor is reversible, with no conformational

change and signal transmission, and the platelet function in

blood also recovers rapidly after drug withdrawal

Cilostazol By inhibiting phosphodiesterase activity in platelets and Cilostazol can be used in AIS patients and as an alternative to aspirin

vascular smooth muscle, the cAMP concentration in platelets

and smooth muscle is increased to give play to the antiplatelet

effect and vasodilation effect. It inhibits platelet aggregation

and release reactions induced by ADP, epinephrine, collagen

and arachidonic acid, and has obvious antithrombotic effect on

models of cerebral circulation and peripheral circulation

disorders induced by collagen, ADP, arachidonic acid and

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 293

sodium laurate

Indobufen It reversibly inhibits COX-1, inhibits platelet aggregation For patients with aspirin intolerance (with gastrointestinal reaction or allergy,

induced by ADP, epinephrine, collagen and arachidonic acid, etc.) and a high risk of haemorrhage, it is feasible to use indobufen (100mg/

and exerts antiplatelet effect. In addition, it can also reduce time, bid)

platelet factor 3 and platelet factor 4, significantly inhibit

coagulation factor II and coagulation factor X, and has

anticoagulant effect

Dipyridamole It inhibits platelet uptake of adenosine, inhibits Whether dipyridamole antithrombotic monotherapy is more beneficial to

phosphodiesterase and increases platelet cAMP, and is a atrong secondary prevention of cerebral infarction still needs to be confirmed by a

agonist that inhibits TXA2 formation and TXA2 platelet large number of RCTs

activity, and enhances endogenous (PGI2)

Abciximab Namely the "anti-platelet aggregation monoclonal antibody", It may be harmful to AIS treatment

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 294

which can selectively block platelet glycoprotein Ⅱb/Ⅲa

receptor and prevent fibrinogen, platelet-activating factor

(PAF), vitreous binding protein and fibrin binding protein from

binding to activated platelets

Tirofiban It is a reversible non-peptide platelet surface GPⅡb/Ⅲa The efficacy and safety have not yet been determined and need further

receptor antagonist. It competitively inhibits the binding of confirmation

fibrinogen to platelet GPⅡb/Ⅲa receptor, inhibits platelet

aggregation, prolongs bleeding time, and inhibits thrombosis

Eptifibatide It selectively blocks the binding of adhesion protein to The efficacy and safety have not yet been determined and need further

GPⅡb/Ⅲa, and is also a relatively weak inhibitor of most confirmation

related receptors

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 295

Supplemental Table 9 Type of haemorrhagic transformation in ischemic stroke

Type Definition Scanning time point

Asymptomatic Imaging examination shows intracranial

intracranial haemorrhage, but there is no neurological

haemorrhage deterioration in the patient

Symptomatic

intracranial

haemorrhage

NINDS CT finds haemorrhage + any neurological 24 hours after onset, 7-10

function decline days after onset or when

clinical symptoms suggest

haemorrhage

ECASS Ⅱ Any haemorrhage + NIHSS score increase 22-36h and 7d after

≥ 4 points treatment or when clinical

symptoms aggravate

SITS-MOST Hematoma volume at infarction site or 22-36h after treatment

distant site > 30% of infarction volume,

with obvious mass effect + NIHSS score

increase ≥ 4 points

ECASS Ⅲ Any haemorrhage + NIHSS score increase 22-36h after CT or MRI

≥ 4 points, and it is determined that treatment

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 296

haemorrhage is the main cause of

neurological deterioration

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 297

Supplemental Table 10 Essen stroke risk score

Risk factors Score/points Risk factors Score/points

Aged 65~75 years 1 Other cardiovascular diseases 1

Age > 75 years 2 (except for myocardial

infarction and atrial

fibrillation)

Hypertension 1 Peripheral arterial disease 1

Diabetes 1 Smoking 1

History of 1 History of TIA or ischemic 1

myocardial stroke

infarction

Total score 9

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 298

Supplemental Table 11 CHADS2 score and CHA2DS2-VASc score

CHADS2 risk factors Score/points CHA2 DS2-VAScrisk Score/points

factors

Heart failure 1 Heart failure 1

Hypertension 1 Hypertension 1

Age > 75 years 1 Age > 75 years 2

Diabetes 1 Diabetes 1

History of stroke/TIA 2 History of stroke/TIA 2

Peripheral vascular 1

disease

Aged 65~74 years 1

Female 1

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 299

Supplemental Table 12 Stratified assessment of stroke risk for atrial fibrillation

patients

CHADS2 Low risk Intermediate risk High risk

Classic CHADS2 score 0 1~2 3~6

Modified CHADS2 score 0 1 2~6

CHA2DS2-VASc score 0 1 2~9

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 300

Supplemental Table 13 HAS-BLED scale

Risk factors Score Risk factors Score

Hypertension 1 Labile INR 1

Abnormal liver 1/2 (1 point for liver Elderly (age > 65 1

and/or kidney and 1 point for kidney) years)

function

History of 1 Drugs and/or 1/2 (1 point for drugs and 1

stroke alcohol point for alcohol)

History or 1

tendency of

haemorrhage

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 301

Supplemental Table 14 SPI-Ⅱ scale

Item Score/points Item Score/points

Age > 70 years 2 Coronary atherosclerotic heart 1

disease

SBP > 180mmHg or DBP > 1 History of stroke 3

100mmHg

Diabetes 3 Congestive cardiac failure 3

Stroke events (non-transient 2

ischemic attack)

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 302

Supplemental Table 15 Comparison table of doses of lipid-lowering drugs

Ultra-high Low intensity Moderate intensity intensity lipid- lipid-lowering lipid-lowering High intensity lipid- lowering treatment treatment lowering treatment treatment Lower LDL-C < Lower LDL-C Lower LDL-C 50%-60% Lower LDL-C > 30% 30%-49% 60%

Atorvastatin 40- Atorvastatin 10- Simvastatin 10mg Atorvastatin 40-80mg 80mg + Ezetimibe 20mg 10mg

Rosuvastatin 20-

Vastatin 10-20mg Rosuvastatin 5-10mg Rosuvastatin 20-40mg 40mg + ezetimibe

10mg

Lovastatin 10- Simvastatin 20- Simvastatin 20-40mg +

20mg 40mg Ezetimibe 10mg

Pravastatin 40mg + Fluvastatin 40mg Pravastatin 40mg Ezetimibe 10mg

Lovastatin 40mg + Pitavastatin 1mg Lovastatin 40mg Ezetimibe 10mg

Fluvastatin 80mg + Ezetimibe 10mg Fluvastatin XL 80mg Ezetimibe 10mg

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378 Supplementary material Stroke Vasc Neurol

Supplemental Material Liu Page 303

Pitavastatin 2-4mg + Pitavastatin 2-4mg Ezetimibe 10mg

Simvastatin 10mg + Atorvastatin 10-20mg +

Ezetimibe 10mg Ezetimibe 10mg

Pravastatin 20mg + Rosuvastatin 5-10mg +

Ezetimibe 10mg Ezetimibe 10mg

Lovastatin 20mg +

Ezetimibe 10mg

Fluvastatin 40mg +

Ezetimibe 10mg

Pitavastatin 1mg +

Ezetimibe 10mg

Liu L, et al. Stroke Vasc Neurol 2020; 5:e000378. doi: 10.1136/svn-2020-000378